US7939527B2 - Thiazolopyridin-2-yloxy-phenyl and thiazolopyrazin-2-yloxy-phenyl amines as modulators of leukotriene A4 hydrolase - Google Patents

Thiazolopyridin-2-yloxy-phenyl and thiazolopyrazin-2-yloxy-phenyl amines as modulators of leukotriene A4 hydrolase Download PDF

Info

Publication number
US7939527B2
US7939527B2 US12/421,406 US42140609A US7939527B2 US 7939527 B2 US7939527 B2 US 7939527B2 US 42140609 A US42140609 A US 42140609A US 7939527 B2 US7939527 B2 US 7939527B2
Authority
US
United States
Prior art keywords
thiazolo
pyridin
phenoxy
yloxy
ethyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
US12/421,406
Other languages
English (en)
Other versions
US20090258854A1 (en
Inventor
Genesis M. Bacani
Diego Broggini
Eugene Y. Cheung
Christa C. Chrovian
Xiaohu Deng
Laurent Gomez
Cheryl A. Grice
Aaron M. Kearney
Adrienne M. Landry-Bayle
Alice Lee-Dutra
Jimmy T. Liang
Susanne Lochner
Neelakandha S. Mani
Alejandro Santillán, Jr.
Kathleen Sappey
Kia Sepassi
Virginia M. Tanis
Alvah T. Wickboldt
John J. M. Wiener
Hartmut Zinser
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Janssen Pharmaceutica NV
Original Assignee
Janssen Pharmaceutica NV
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to US12/421,406 priority Critical patent/US7939527B2/en
Application filed by Janssen Pharmaceutica NV filed Critical Janssen Pharmaceutica NV
Assigned to JANSSEN PHARMACEUTICA NV reassignment JANSSEN PHARMACEUTICA NV ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GRICE, CHERYL A.
Assigned to JANSSEN PHARMACEUTICA NV reassignment JANSSEN PHARMACEUTICA NV ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BACANI, GENESIS M., CHROVIAN, CHRISTA C., DENG, XIAOHU, GOMEZ, LAURENT, KEARNEY, AARON M., LANDRY-BAYLE, ADRIENNE M., LEE-DUTRA, ALICE, LIANG, JIMMY T., MANI, NEELAKANDHA S., SANTILLAN, JR, ALEJANDRO, SAPPEY, KATHLEEN C., SEPASSI, KIA, TANIS, VIRGINIA M., WIENER, JOHN J.M.
Assigned to JANSSEN PHARMACEUTICA NV reassignment JANSSEN PHARMACEUTICA NV ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BROGGINI, DIEGO, LOCHNER, SUSANNE, ZINSER, HARTMUT
Assigned to JANSSEN PHARMACEUTICA NV reassignment JANSSEN PHARMACEUTICA NV ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHEUNG, EUGENE Y.
Assigned to JANSSEN PHARMACEUTICA NV reassignment JANSSEN PHARMACEUTICA NV ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: FOURIE, ANNE M.
Assigned to JANSSEN PHARMACEUTICA NV reassignment JANSSEN PHARMACEUTICA NV ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: WICKBOLDT, ALVAH T.
Publication of US20090258854A1 publication Critical patent/US20090258854A1/en
Priority to US13/039,017 priority patent/US8357684B2/en
Priority to US13/039,105 priority patent/US20110190503A1/en
Publication of US7939527B2 publication Critical patent/US7939527B2/en
Application granted granted Critical
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/16Otologicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/12Drugs for disorders of the metabolism for electrolyte homeostasis
    • A61P3/14Drugs for disorders of the metabolism for electrolyte homeostasis for calcium homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/38Drugs for disorders of the endocrine system of the suprarenal hormones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D451/00Heterocyclic compounds containing 8-azabicyclo [3.2.1] octane, 9-azabicyclo [3.3.1] nonane, or 3-oxa-9-azatricyclo [3.3.1.0<2,4>] nonane ring systems, e.g. tropane or granatane alkaloids, scopolamine; Cyclic acetals thereof
    • C07D451/02Heterocyclic compounds containing 8-azabicyclo [3.2.1] octane, 9-azabicyclo [3.3.1] nonane, or 3-oxa-9-azatricyclo [3.3.1.0<2,4>] nonane ring systems, e.g. tropane or granatane alkaloids, scopolamine; Cyclic acetals thereof containing not further condensed 8-azabicyclo [3.2.1] octane or 3-oxa-9-azatricyclo [3.3.1.0<2,4>] nonane ring systems, e.g. tropane; Cyclic acetals thereof

Definitions

  • the present invention relates to certain thiazolopyridin-2-yloxy-phenyl and thiazolopyrazin-2-yloxy-phenyl amine compounds, pharmaceutical compositions containing them, and methods of using the compounds and pharmaceutical compositions for leukotriene A4 hydrolase (LTA4H) modulation and for the treatment of disease states, disorders, and conditions mediated by leukotriene A4 hydrolase (LTA4H) activity.
  • LTA4H leukotriene A4 hydrolase
  • Inflammation is normally an acute response by the immune system to invasion by microbial pathogens, chemicals or physical injury. In some cases, however, the inflammatory response can progress to a chronic state, and be the cause of inflammatory disease. Therapeutic control of this chronic inflammation in diverse diseases is a major medical need.
  • Leukotrienes are biologically active metabolites of arachidonic acid (B. Samuelsson, Science 1983, 220(4597): 568-575) that have been implicated in inflammatory diseases, including asthma (D. A. Munafo et al., J. Clin. Invest. 1994, 93(3): 1042-1050; N. Miyahara, et al., Allergol Int., 2006, 55(2): 91-7; E. W. Gelfand, et al., J. Allergy Clin. Immunol. 2006, 117(3): 577-82; K. Terawaki, et al., J. Immunol.
  • IBD inflammatory bowel disease
  • COPD chronic obstructive pulmonary disease
  • arthritis R. J. Griffiths et al., Proc. Natl. Acad. Sci. U.S.A. 1995, 92(2): 517-521; F. Tsuji et al., Life Sci. 1998, 64(3): L51-L56
  • psoriasis K. Ikai, J. Dermatol. Sci. 1999, 21(3): 135-146; Y. I.
  • leukotriene A4 leukotriene A4
  • 5-lipoxygenase A. W. Ford-Hutchinson et al., Annu. Rev. Biochem. 1994, 63: 383-347.
  • This enzyme is expressed predominantly by cells of myeloid origin, particularly neutrophils, eosinophils, monocytes/macrophages and mast cells (G. K. Reid et al., J. Biol. Chem. 1990, 265(32): 19818-19823).
  • LTA4 can either be conjugated with glutathione by leukotriene C4 (LTC4) synthase to produce the cysteinyl leukotriene, LTC4, or hydrolyzed to the diol, leukotriene B4 (LTB4) (B. Samuelsson, Science 1983, 220(4597): 568-575).
  • LTC4 and its metabolites, LTD4 and LTE4 induce smooth muscle contraction, broncho-constriction and vascular permeability, while LTB4 is a potent chemo-attractant and activator of neutrophils, eosinophils, monocytes/macrophages, T cells and mast cells.
  • LTA4H leukotriene A4 hydrolase
  • This enzyme is ubiquitously expressed, with high levels in small intestinal epithelial cells, lung, and aorta (B. Samuelsson and C. D. Funk, J. Biol. Chem. 1989, 264(33): 19469-19472).
  • Moderate expression of LTA4H is observed in leukocytes, particularly neutrophils (T. Yokomizo et al., J. Lipid Mediators Cell Signalling 1995, 12(2,3): 321-332).
  • Leukotriene B4 is a key pro-inflammatory lipid mediator, able to recruit and activate inflammatory cells, such as neutrophils, eosinophils, monocytes/macrophages, T cells and mast cells (F. A. Fitzpatrick et al., Ann. N.Y. Acad. Sci. 1994, 714: 64-74; S. W. Crooks and R. A. Stockley, Int. J. Biochem. Cell Biol. 1998, 30(2): 173-178; A. Klein et al., J. Immunol. 2000, 164: 4271-4276).
  • inflammatory cells such as neutrophils, eosinophils, monocytes/macrophages, T cells and mast cells
  • LTB4 mediates its pro-inflammatory effects by binding to G protein-coupled receptors, leukotriene B4 receptor 1 (BLT1) and leukotriene B4 receptor 2 (BLT2) (T. Yokomizo et al., Arch. Biochem. Biophys. 2001, 385(2): 231-241).
  • BLT1 leukotriene B4 receptor 1
  • BLT2 leukotriene B4 receptor 2
  • the receptor first identified, BLT1 binds LTB4 with high affinity, leading to intracellular signaling and chemotaxis.
  • BLT1 is expressed mainly in peripheral leukocytes, particularly neutrophils, eosinophils, macrophages (Huang, W. W. et al. J Exp Med 188, 1063-74 (1998)) and monocytes (Yokomizo, T., Izumi, T.
  • the murine receptor is also expressed on effector T cells and was recently shown to mediate LTB4-dependent migration of effector CD8 + T cells (Goodarzi, K., Goodarzi, M., Tager, A. M., Luster, A. D. & von Andrian, U. H. Nat Immunol 4, 965-73 (2003); Ott, V. L., Cambier, J. C., Kappler, J., Marrack, P. & Swanson, B. J.
  • BLT2 binds LTB4 with lower affinity than BLT1 does, mediates chemotaxis at higher concentrations of LTB4, and differs from BLT1 in its affinity for certain antagonists. While LTB4 receptor antagonists may differ in their affinity for BLT1 versus BLT2, blocking the production of LTB4 using LTA4H inhibitors would be expected to inhibit the downstream events mediated through both BLT1 and BLT2.
  • inhibitors of LTB4 production should have therapeutic value as anti-inflammatory agents for these conditions.
  • reduction of LTB4 production by an inhibitor of LTA4H activity would be predicted to have therapeutic potential in a wide range of diseases.
  • LTA4H inhibitors have been shown to be effective anti-inflammatory agents in pre-clinical studies.
  • oral administration of LTA4H inhibitor SC57461 caused inhibition of ionophore-induced LTB4 production in mouse blood ex vivo, and in rat peritoneum in vivo (J. K. Kachur et al., J. Pharm. Exp. Ther.
  • inflammatory diseases or inflammation-mediated diseases or conditions include, but are not limited to, acute inflammation, allergic inflammation, and chronic inflammation.
  • Atopic dermatitis is a chronic inflammatory skin disease that usually occurs in individuals with a personal or family history of atopy. The major features are pruritus and chronic or relapsing eczematous lesions. Complications include bacterial, fungal and viral infections as well as ocular disease. Atopic dermatitis is the most common inflammatory skin disease in children and affects more than 15% of children in the US (Laughter, D., et al., J. Am. Acad. Dermatol. 2000, 43, 649-655). Atopic dermatitis may persist in 60% of adults who were affected as children (Sidbury, R., et al., Dermatol. Clin. 2000, 18(1), 1-11).
  • Atopic dermatitis has significant societal impact.
  • the family stress related to caring for children with moderate to severe AD may be comparable to the stress seen in families of children with type I diabetes mellitus (Su, J. C., et al., Arch. Dis. Child 1997, 76, 159-162).
  • the annual cost of medical services and prescription drugs for the treatment of AD/eczema is similar to those for emphysema, psoriasis and epilepsy (Ellis, C. N., et al., J. Am. Acad. Dermatol. 2002, 46, 361-370).
  • LTB4 levels are elevated in skin lesions (K. Fogh et al., J. Allergy Clin. Immunol. 1989, 83, 450-455; T. Ruzicka et al., J. Invest. Dermatol. 1986, 86, 105-108) and plasma in AD, and contribute to the inflammation through chemotactic effects on inflammatory cells (Wedi and Kapp BioDrugs. 2001; 15, 729-743. Reported in vivo and in vitro studies have shown that leukotrienes, especially LTB4, contribute to the inflammation of the skin in AD through their chemotactic effect on inflammatory cells.
  • LTB4 receptors are expressed on mast cells, T cells, eosinophils, dendritic cells and macrophages, all of which accumulate in AD lesions.
  • LTB4 itself is a pruritic agent, and has also been shown to mediate substance P-induced pruritus (T. Andoh et al., J. Invest. Dermatol. 2001, 117, 1621-1626), a key component of the itching in AD (T. Ohmura et al., Eur. J. Pharmacol. 2004, 491, 191-194).
  • LTB4 induces proliferation of keratinocytes, an effect that is further potentiated by substance P (M. J. Rabier et al., J. Invest. Dermatol.
  • LTB4 in development of a Th2 immune response and IgE production.
  • the role of LTB4 in AD is supported by beneficial effects of the 5-lipoxygenase inhibitor, zileuton, in a small, open-label clinical trials of AD (Woodmansee, D. P., et al., Ann. Allergy Asthma Immunol. 1999, 83, 548-552) and in relieving the pruritus in Sjogren-Larsson syndrome patients who have elevated LTB4 due to an impairment in its degradation (Willemsen, M. A., et al., Eur. J. Pediatr. 2001, 160, 711-717).
  • Emollients have a steroid-sparing effect and are useful for both prevention and maintenance therapy.
  • Crude coal tar and preparations containing coal tar derivatives have also been used for many years in the treatment of AD and have significant cosmetic disadvantages that influence compliance (Hanifin, et al., 2004).
  • Topical doxepin may be a useful short-term adjunctive therapy for the relief of pruritus but sedation and contact dermatitis may complicate its use (Hanifin, et al., 2004).
  • the topical calcineurin inhibitors tacrolimus (Protopic®) and pimecrolimus (Elidel®) have been shown to reduce the extent, severity and symptoms of AD in adults and children and are approved for use as second-line therapy of AD.
  • the recent addition of boxed warnings to the product labels regarding rare cases of malignancy reported in patients treated with topical calcineurin inhibitors limits long term use of these agents in the treatment of AD (Food and Drug Administration [FDA]/Center for Drug Evaluation and Research [CDER] resources page).
  • Antibiotics are used in the treatment of Staphylococcus aureus infections in patients with AD but have a minimal effect on the dermatitis (Hanifin, et al., 2004). Although sedating antihistamines may be useful if sleep disruption is present, oral antihistamines are generally not effective in treating AD-associated pruritus (Hanifin, et al., 2004). Ultraviolet (UV) phototherapy, including photochemotherapy with psoralen is well established in the treatment of AD but relapse upon cessation of therapy frequently occurs (Hanifin, et al., 2004).
  • UV phototherapy including photochemotherapy with psoralen is well established in the treatment of AD but relapse upon cessation of therapy frequently occurs (Hanifin, et al., 2004).
  • Systemic immunomodulatory therapy with cyclosporine and corticosteroids is effective but can be associated with severe side effects and is generally reserved for patients with severe disease.
  • Systemic corticosteroids are associated with growth retardation in children, avascular necrosis of bone, osteopenia, increased risk of infection, poor wound healing, cataracts, hyperglycemia and hypertension.
  • Cyclosporine is nephrotoxic in a majority of patients and is associated with tremor, hirsutism, hypertension, hyperlipidemia and gum hyperplasia.
  • AD While AD that is mild to moderate in severity generally responds to topical therapy, correct use of these therapies and compliance remain a major issue in the clinic.
  • An effective oral or topical therapy with fewer side effects than systemic immunomodulatory therapies and potent topical corticosteroids would fill an unmet medical need in the treatment of AD.
  • Sjogren-Larsson syndrome is an autosomal recessive neurocutaneous disorder with severe ichthyosis. It is caused by mutation of the gene encoding microsomal fatty aldehyde dehydrogenase (FALDH) leading to a defect in fatty alcohol metabolism. FALDH catalyzes the oxidation of medium- to long-chain fatty aldehydes to their corresponding carboxylic acids. LTB4, a pro-inflammatory mediator synthesized from arachidonic acid, is inactivated by microsomal omega-oxidation, successively yielding 20-OH-LTB4, 20-CHO-LTB4 and 20-COOH-LTB4.
  • FALDH microsomal fatty aldehyde dehydrogenase
  • Sjogren-Larsson syndrome is an autoimmune disease that features inflammation in some glands. Sjogren syndrome may feature also extraglandular manifestations. When the gland inflammation is not associated with another connective tissue disease, then the syndrome is referred to as primary Sjogren syndrome.
  • Sjogren syndrome When it is associated with a connective tissue disease, such as rheumatoid arthritis, systemic lupus erythematosus or scleroderma, then it is referred to as secondary Sjogren syndrome.
  • the term “Sjogren syndrome” herein refers to any one of the primary and secondary Sjogren syndromes. No cure is currently known for this syndrome.
  • the current treatments usually focus on the specific area of the body that is affected and also in the treatment of associated complications.
  • Immuno-suppressants such as cortisones, azathioprine and cyclophosphamide are sometimes used to threat some serious complications, and antibiotics are also used to treat associated infections.
  • Embodiments of this invention have shown dose-dependent inhibition of dermal inflammation in the arachidonic acid-induced murine ear inflammation model. Oral administration of embodiments of this invention dose-dependently inhibited neutrophil influx and edema, and were shown to inhibit the ex vivo ionophore-stimulated LTB4 production at doses between 0.3 and 30 mg/kg.
  • LTA4H inhibitors are hypothesized to specifically block the production of LTB4 from LTA4, without affecting the biosynthesis of lipoxins, which are also produced from LTA4.
  • Increasing or maintaining lipoxin A 4 (LXA 4 ) production may have beneficial therapeutic effects in dermal inflammation as it has been reported that topical application of a stable lipoxin analogue inhibits edema, granulocyte infiltration and epidermal hyperproliferation in murine skin inflammation models.
  • 5-LO inhibitors block the pathway upstream of LTA4. This would be expected to lead to a block in not only synthesis of LTA4, LTB4 and cysteinyl leukotrienes (CysLT), but also LXA 4 .
  • Embodiments of this invention have been studied in in vivo inflammation models including arachidonic acid-induced ear inflammation and allergic lung inflammation, including an ovalbumin (OVA) sensitization and airway challenge model and a rat ionophore-induced lung inflammation model.
  • OVA ovalbumin
  • embodiments of the invention show dose-dependent inhibition of the accumulation of inflammatory cells in the lungs in an ovalbumin-induced allergic airway inflammation model, a common animal model for human allergic inflammation.
  • Asthma is a chronic disease characterized by a variable degree of airflow obstruction, bronchial hyperresponsiveness and airway inflammation (Busse & Lemanske, 2001).
  • Immunohistopathologic features include denudation of airway epithelium, collagen deposition beneath basement membrane, edema, mast cell activation, and inflammatory cell infiltration by neutrophils (especially in sudden-onset, fatal asthma exacerbations), eosinophils, and Th2 lymphocytes (W. W. Busse et al., N. Engl. J. Med. 2001, 344, 350-362).
  • Airway inflammation contributes to the airway hyperresponsiveness, airflow limitation (acute bronchoconstriction, airway edema, mucus plug formation and airway wall remodeling, leading to bronchial obstruction), respiratory symptoms and disease chronicity (NIH Guidelines for the Diagnosis and Management of Asthma 1997).
  • Glanal asthma Current therapy for asthma is directed at controlling acute bronchoconstrictive symptoms with beta2-adrenergic receptor agonists and managing underlying airway inflammation with inhaled corticosteroids, chromates such as cromolyn sodium and nedocromil, and antileukotriene agents, such as the cysteinyl leukotriene receptor antagonists montelukast and zafirlukast and the 5-lipoxygenase inhibitor zileuton.
  • Systemic steroids are used in severe disease and acute exacerbations of asthma.
  • the humanized monoclonal anti-IgE antibody omalizumab was approved for the treatment of patients with moderate-to-severe persistent asthma who have a positive skin test or in vitro reactivity to a perennial aeroallergen and whose symptoms are inadequately controlled with inhaled corticosteroids (XOLAIR® [omalizumab] July 2007).
  • the inflammatory component of mild persistent and moderate asthma can generally be controlled with inhaled corticosteroids, but patient compliance remains a major issue in disease management (H. Milgrom et al., J. Allergy Clin. Immunol. 1996, 98, 1051-1057).
  • patient compliance remains a major issue in disease management (H. Milgrom et al., J. Allergy Clin. Immunol. 1996, 98, 1051-1057).
  • many patients have poorly controlled asthma (J. M. Fitzgerald et al., Can. Respir. J. 2006, 13, 253-259; D. Bellamy et al., Prim. Care Respir. J. 2005, 14, 252-258).
  • Severe asthma requires treatment with high-dose inhaled steroids or the frequent use of oral corticosteroids (W. Moore et al., J.
  • Leukotrienes are important mediators in asthma pathogenesis and comprise two classes—the cysteinyl leukotrienes (LTC4, LTD4 and LTE4) and LTB4.
  • Leukotriene receptor antagonists such as montelukast or zafirlukast, target only the cysteinyl leukotrienes, while 5-lipoxygenase inhibitors, such as zileuton, inhibit the pathway upstream of both classes, and thus decrease formation of both the cysteinyl leukotrienes and LTB4.
  • LTA4H inhibitors selectively inhibit LTB4 synthesis and do not impact cysteinyl leukotriene (CysLT) synthesis. Both classes of leukotrienes are elevated in asthma, and LTB4 is more significantly increased in severe asthma, which is associated with increased neutrophilic inflammation.
  • LTB4 plays a role in eosinophil and effector T cell recruitment, IL-13 production, goblet cell hyperplasia and mucus secretion, IgE production and airway hyperreactivity (Miyahara et al., Allergy Intl. 2006, 55, 91-97).
  • LTB4 receptor antagonist CP-105,696
  • Allergic rhinitis is an inflammation of the mucus membranes of the nose that occurs in response to an airborne antigen (allergen).
  • Allergic rhinitis also called allergic rhinoconjunctivitis, is characterized by frequent or repetitive sneezing, runny or congested nose, and pruritus of the nose, eyes and throat. It may also be associated with other symptoms such as headache, impaired smell, postnasal drip, conjunctival symptoms (e.g., itchy watery eyes), sinusitis and other complicating respiratory symptoms.
  • allergic rhinitis can be classified as perennial, seasonal or occupational.
  • Embodiments of this invention have shown dose-dependent inhibition of lung inflammation in pre-clinical models, Based upon the well-described leukotriene biosynthesis pathway, LTA4H inhibitors are hypothesized to specifically block the production of LTB4 from LTA4, without affecting the biosynthesis of lipoxins, which are also produced from LTA4.
  • Lipoxins such as LXA 4
  • LXA 4 have been the focus of intense study and are known to play a key role as natural anti-inflammatory agents and key mediators of the natural process of resolving an inflammatory response.
  • production of endogenous LXA 4 has been described in a variety of inflammatory diseases and lower levels of LXA 4 have been found in patients with severe versus moderate asthma.
  • LXA 4 plays an important role in resolution of acute inflammation.
  • 5-LO inhibitors block this pathway upstream of LTA4. This would lead to a block in not only synthesis of LTA4, LTB4 and cysteinyl leukotrienes, but also LXA 4 .
  • LTA4H inhibitors result in a buildup of LTA4, and pathway shunting to pro-inflammatory cysteinyl leukotrienes, although to date there is no known data to support this possibility.
  • Neutrophil infiltration is a prominent feature of severe asthma.
  • Zileuton Zyflo®
  • CysLT antagonists for example, Montelukast/Singulair®
  • Combination of an LTA4H inhibitor and at least one of a CysLT receptor antagonist and LTC4 synthase inhibitor would target both LTB4 and cysteinyl leukotrienes, while leaving production of the anti-inflammatory lipoxins intact.
  • Embodiments of this invention reduced inflammatory responses to airway allergen challenge in sensitized mice, leading to dose-dependent decreases airway recruitment of inflammatory cells.
  • Embodiments of this invention are expected to find utility in treating inflammatory bowel disease.
  • TNBS trinitrobenzene sulfonic acid
  • LTA4H inhibition had significant inhibitory effects on colonic inflammation, including macroscopic colonic injury, inflammatory cell content, and levels of tumor necrosis factor alpha (TNF- ⁇ ), LTB4, and IL-6.
  • TNF- ⁇ tumor necrosis factor alpha
  • LTA4H inhibition also significantly attenuated the joint inflammation and swelling associated with the destruction of collagen in murine models of arthritis. Mice deficient in receptors for LTB4 or lacking LTA4H do not develop arthritis in murine models (Mathis, S., et al.
  • Embodiments of this invention are thus expected to find utility in treating arthritis, including, but not limited to, rheumatoid arthritis.
  • Abdominal aortic aneurysm is a localized dilatation of the abdominal aorta that exceeds the normal diameter (2 cm) by more than 50%. It is caused by a degenerative process of the aortic wall. An aortic aneurysm may also occur in the thorax. Surgery is eventually required to prevent the progression to AAA rupture, which is most often a fatal event. Thus therapeutics which delay or prevent the need for surgery are an unmet medical need.
  • LTB4-receptor-deficient mice Diminished AAA formation in LTB4-receptor-deficient mice was associated with significant reductions in mononuclear cell chemoattractants and leukocyte accumulation in the vessel wall, as well as striking reductions in the production of matrix metalloproteinases-2 and -9.
  • signaling by LTB4 through its receptor contributes to the frequency and size of abdominal aortic aneurysms in mice, and prevention of LTB4 signaling by deletion of the gene coding for the LTB4 receptor in turn inhibits proinflammatory circuits and enzymes that modulate vessel wall integrity.
  • LTB4 signaling is a target for intervention in modulating development of aortic aneurysms.
  • Inhibitors of LTA4H in the context of this invention are expected to have utility in inhibition of aortic aneurysms.
  • Embodiments of this invention are expected to find utility in treating also any one or a combination of atopic dermatitis, contact dermatitis, acne (T. Alestas, et al., J. Mol. Med. 2006, 84(1): 75-87; Ch. C. Zouboulis, et al., Dermatology, 2005, 210(1): 36-8; Arch. Dermatol. 2003, 139(5): 668-70), myocardial infarction (A. Helgadottir, et al., Nat. Genet. 2006, 38(1): 68-74; Nat. Genet. 2004, 36(3): 233-9; H.
  • Inflammation is due to or associated with any one of a plurality of conditions, such as asthma, chronic obstructed pulmonary disease (COPD), atherosclerosis, rheumatoid arthritis, multiple sclerosis, inflammatory bowel diseases (including Crohn's disease and ulcerative colitis), psoriasis, atopic dermatitis, contact dermatitis, acne, myocardial infarction, stroke, pain, itch (pruritus), gingivitis, uveitis, bronchitis, allergic rhinitis, cystic fibrosis, upper gastrointestinal cancer, sepsis, Sjogren syndrome, Sjogren-Larssen syndrome, and skin burns, which are each characterized by excessive or prolonged inflammation at some stage of the disease.
  • COPD chronic obstructed pulmonary disease
  • atherosclerosis rheumatoid arthritis
  • multiple sclerosis multiple sclerosis
  • inflammatory bowel diseases including Crohn's disease and ulcerative colitis
  • Organ transplant rejection and autoimmune disease treatment with a cyclooxygenase-2 inhibitor and an LTA4H inhibitor are disclosed in WO1997/29774, U.S. Patent Appl. Publ. Nos. US2003/004191 and US2005/043355, and in U.S. Pat. Nos. 5,700,816, 6,407,140.
  • LTA4H inhibitors are disclosed in U.S. Pat. Nos. 5,719,306, 6,506,876, 5,723,492, 5,585,492, and publication WO1996/11192.
  • Cyclic and bicyclic diamino histamine-3 receptor antagonists are disclosed in U.S. Pat. No. 6,559,140.
  • Patent Appl. Publ. No. WO2007/079078 Aryl-substituted bridged diamines are disclosed as LTA4H modulators in U.S. Provisional Pat. Appl. No. 60/984,126. Combinations of a cyclooxygenase-2 inhibitor and an LTA4H inhibitor for the treatment of inflammation and inflammation-related disorders are disclosed in U.S. Pat. No. 5,990,148 and in publication WO1996/41625. Nitrogeneous derivatives have been disclosed in patent-related as well as in nonpatent-related publications, such as WO2008/016811; US2008/0057074; WO2006/002133; U.S. Pat. No. 6,316,490; U.S. Pat. No.
  • the invention relates to chemical entities selected from compounds of Formula (I), pharmaceutically acceptable salts of compounds of Formula (I), pharmaceutically acceptable prodrugs of compounds of Formula (I), solvates of compounds of Formula (I), and pharmaceutically active metabolites of compounds of Formula (I):
  • the compound of Formula (I) is a compound selected from those species described or exemplified in the detailed description below.
  • compositions each comprising an effective amount of at least one chemical entity selected from compounds of Formula (I), pharmaceutically acceptable salts of compounds of Formula (I), pharmaceutically acceptable prodrugs of compounds of Formula (I), solvates of compounds of Formula (I), and pharmaceutically active metabolites of Formula (I).
  • Pharmaceutical compositions according to the invention may further comprise a pharmaceutically acceptable excipient.
  • embodiments of the invention are useful as LTA4H modulators.
  • the invention is directed to a method for modulating LTA4H activity, comprising exposing LTA4H to an effective amount of at least one chemical entity selected from compounds of Formula (I), pharmaceutically acceptable salts of compounds of Formula (I), pharmaceutically acceptable prodrugs of compounds of Formula (I), and pharmaceutically active metabolites of compounds of Formula (I).
  • Embodiments of this invention inhibit LTA4H activity.
  • the invention is directed to a method of treating a subject suffering from or diagnosed with a disease, disorder, or medical condition mediated by LTA4H activity, comprising administering to the subject in need of such treatment an effective amount of at least one chemical entity selected from compounds of Formula (I), pharmaceutically acceptable salts of compounds of Formula (I), pharmaceutically acceptable prodrugs of compounds of Formula (I), and pharmaceutically active metabolites of compounds of Formula (I).
  • the disease, disorder, or medical condition is inflammation, atopic dermatitis, or asthma.
  • An object of the present invention is to overcome or ameliorate at least one of the disadvantages of the conventional methodologies and/or prior art, or to provide a useful alternative thereto.
  • alkyl refers to a straight- or branched-chain alkyl group having from 1 to 12 carbon atoms in the chain.
  • alkyl groups include methyl (Me), ethyl (Et), n-propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl (tBu), pentyl, isopentyl, tert-pentyl, hexyl, isohexyl, and groups that in light of the ordinary skill in the art and the teachings provided herein would be considered equivalent to any one of the foregoing examples.
  • cycloalkyl refers to a saturated or partially saturated, monocyclic, fused polycyclic, or spiro polycyclic carbocycle having from 3 to 12 ring atoms per carbocycle.
  • Illustrative examples of cycloalkyl groups include the following entities, in the form of properly bonded moieties:
  • heterocycloalkyl refers to a monocyclic, or fused, bridged, or spiro polycyclic ring structure that is saturated or partially saturated and has from 3 to 12 ring atoms per ring structure selected from carbon atoms and up to three heteroatoms selected from nitrogen, oxygen, and sulfur.
  • the ring structure may optionally contain up to two oxo groups on carbon or sulfur ring members.
  • Illustrative entities, in the form of properly bonded moieties include:
  • heteroaryl refers to a monocyclic, fused bicyclic, or fused polycyclic aromatic heterocycle (ring structure having ring atoms selected from carbon atoms and up to four heteroatoms selected from nitrogen, oxygen, and sulfur) having from 3 to 12 ring atoms per heterocycle.
  • heteroaryl groups include the following entities, in the form of properly bonded moieties:
  • halogen represents chlorine, fluorine, bromine, or iodine.
  • halo represents chloro, fluoro, bromo, or iodo.
  • substituted means that the specified group or moiety bears one or more substituents.
  • unsubstituted means that the specified group bears no substituents.
  • optionally substituted means that the specified group is unsubstituted or substituted by one or more substituents. Where the term “substituted” is used to describe a structural system, the substitution is meant to occur at any valency-allowed position on the system.
  • any formula given herein is intended to represent compounds having structures depicted by the structural formula as well as certain variations or forms.
  • compounds of any formula given herein may have asymmetric centers and therefore exist in different enantiomeric forms. All optical isomers and stereoisomers of the compounds of the general formula, and mixtures thereof, are considered within the scope of the formula.
  • any formula given herein is intended to represent a racemate, one or more enantiomeric forms, one or more diastereomeric forms, one or more atropisomeric forms, and mixtures thereof.
  • certain structures may exist as geometric isomers (i.e., cis and trans isomers), as tautomers, or as atropisomers.
  • meso compounds which are compounds that possess asymmetric centers (in this case, asymmetric carbons), but which are achiral molecules. Such compounds are named herein as meso compounds. In some cases, meso compounds are depicted and named herein with a specific stereochemical configuration. However, one skilled in the art will recognize the meso nature of such compounds. Examples include meso-3,8-diazabicyclo[3.2.1]octane-3-carboxamide and meso-1-[(3-endo)-8-azabicyclo[3.2.1]oct-3-yl]urea.
  • any formula given herein is intended to refer also to hydrates, solvates, and polymorphs of such compounds, and mixtures thereof, even if such forms are not listed explicitly.
  • Certain compounds of Formula (I) or pharmaceutically acceptable salts of compounds of Formula (I) may be obtained as solvates.
  • Solvates include those formed from the interaction or complexation of compounds of the invention with one or more solvents, either in solution or as a solid or crystalline form. In some embodiments, the solvent is water and then the solvates are hydrates. Hydrates, such as the monohydrate, of compounds of formula (I) were obtained. Solvates of salts of compounds of formula (I) were obtained in solvated, including hydrated, forms.
  • Solvated salts included, for example, hydrocholorides, phosphates, benzoates, and sulfates. Solvates included hydrates and methanolates. Some embodiments of solvates were mono-solvates, such as monohydrates and monomethanolates. Other embodiments of solvates were hemisolvates, such as hemihydrates.
  • certain crystalline forms of compounds of Formula (I) or pharmaceutically acceptable salts of compounds of Formula (I) may be obtained as co-crystals. In certain embodiments of the invention, compounds of Formula (I) were obtained in a crystalline form. In other embodiments, pharmaceutically acceptable salts of compounds of Formula (I) were obtained in a crystalline form.
  • crystals of compounds of formula (I) and salts thereof exist in a plurality of forms, which can be isolated according to a plurality of methods.
  • crystals of compounds of formula (I) and their salts were obtained by crystallization from a ketone-based medium, such as from 2-butanone, acetone, and a methanol/methyl ethyl ketone mixture.
  • crystals in other forms of compounds of formula (I) and their salts were obtained by crystallization from an acidic medium, such as from methanol with lactic acid.
  • crystals in other forms of compounds of formula (I) and their salts were obtained by crystallization from an acetonitrile-based medium, such as from a methanol/acetonitrile mixture.
  • solvents from which embodiments of salts according to this invention were crystallized include anisole/pyridine mixtures, m-xylene/pyridine mixtures, dimethyl sulfoxide (DMSO)/m-xylene mixtures, methanol/methyl-t-butyl ester (MTBE) mixtures, m-xylene/N-methylpyrrolidone (NMP) mixtures, and chloroform/NMP mixtures.
  • compounds of Formula (I) were obtained in one of several polymorphic forms, as a mixture of crystalline forms, as a polymorphic form, or as an amorphous form. In other embodiments, compounds of Formula (I) convert in solution between one or more crystalline forms and/or polymorphic forms.
  • references to a chemical entity herein stands for a reference to any one of: (a) the actually recited form of such chemical entity, and (b) any of the forms of such chemical entity in the medium in which the compound is being considered when named.
  • reference herein to a compound such as R—COOH encompasses reference to any one of, for example, R—COOH (s) , R—COOH (sol) , and R—COO ⁇ (sol) .
  • R—COOH (s) refers to the solid compound, as it could be for example in a tablet or some other solid pharmaceutical composition or preparation
  • R—COOH (sol) refers to the undissociated form of the compound in a solvent
  • R—COO ⁇ (sol) refers to the dissociated form of the compound in a solvent, such as the dissociated form of the compound in an aqueous environment, whether such dissociated form derives from R—COOH, from a salt thereof, or from any other entity that yields R—COO ⁇ upon dissociation in the medium being considered.
  • an expression such as “exposing an entity to compound of formula R—COOH” refers to the exposure of such entity to the form, or forms, of the compound R—COOH that exists, or exist, in the medium in which such exposure takes place.
  • an expression such as “reacting an entity with a compound of formula R—OOH” refers to the reacting of (a) such entity in the chemically relevant form, or forms, of such entity that exists, or exist, in the medium in which such reacting takes place, with (b) the chemically relevant form, or forms, of the compound R—COOH that exists, or exist, in the medium in which such reacting takes place.
  • a zwitterionic compound is encompassed herein by referring to a compound that is known to form a zwitterion, even if it is not explicitly named in its zwitterionic form.
  • Terms such as zwitterion, zwitterions, and their synonyms zwitterionic compound(s) are standard IUPAC-endorsed names that are well known and part of standard sets of defined scientific names.
  • the name zwitterion is assigned the name identification CHEBI:27369 by the Chemical Entities of Biological merest (ChEBI) dictionary of molecular entities. (See, for example its on line version at http://www.ebi.ac.uk/chebi/init.do).
  • a zwitterion or zwitterionic compound is a neutral compound that has formal unit charges of opposite sign. Sometimes these compounds are referred to by the term “inner salts”. Other sources refer to these compounds as “dipolar ions”, although the latter term is regarded by still other sources as a misnomer.
  • aminoethanoic acid the amino acid glycine
  • H 2 NCH 2 COOH the amino acid glycine
  • it exists in some media in this case in neutral media in the form of the zwitterion + H 3 NCH 2 COO ⁇ .
  • Zwitterions, zwitterionic compounds, inner salts and dipolar ions in the known and well established meanings of these terms are within the scope of this invention, as would in any case be so appreciated by those of ordinary skill in the art. Because there is no need to name each and every embodiment that would be recognized by those of ordinary skill in the art, no structures of the zwitterionic compounds that are associated with the compounds of this invention are given explicitly herein. They are, however, part of the embodiments of this invention. No further examples in this regard are provided herein because the interactions and transformations in a given medium that lead to the various forms of a given compound are known by any one of ordinary skill in the art.
  • any formula given herein is also intended to represent unlabeled forms as well as isotopically labeled forms of the compounds.
  • Isotopically labeled compounds have structures depicted by the formulas given herein except that one or more atoms are replaced by an atom having a selected atomic mass or mass number.
  • isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, chlorine, and iodine, such as 2 H, 3 H, 11 C, 13 C, 14 C, 15 N, 18 O, 17 O, 32 P, 33 P, 35 S, 18 F, 36 Cl, and 125 I, respectively.
  • Such isotopically labelled compounds are useful in metabolic studies (preferably with 14 C), reaction kinetic studies (with, for example 2 H or 3 H), detection or imaging techniques [such as positron emission tomography (PET) or single-photon emission computed tomography (SPECT)] including drug or substrate tissue distribution assays, or in radioactive treatment of patients.
  • PET positron emission tomography
  • SPECT single-photon emission computed tomography
  • an 18 F or 11 C labeled compound may be particularly preferred for PET or SPECT studies.
  • substitution with heavier isotopes such as deuterium (i.e., 2 H) may afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements.
  • Isotopically labeled compounds of this invention and prodrugs thereof can generally be prepared by carrying out the procedures disclosed in the schemes or in the examples and preparations described below by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent.
  • embodiments of this invention comprise the various groupings that can be made from the listed assignments, taken independently, and equivalents thereof.
  • substituent S example is one of S 1 , S 2 , and S 3
  • this listing refers to embodiments of this invention for which S example is S 1 ; S example is S 2 ; S example is S 3 ; S example is one of S 1 and S 2 ; S example is one of S 1 and S 3 ; S example is one of S 2 and S 3 ; S example is one of S 1 , S 2 and S 3 ; and S example is any equivalent of each one of these choices.
  • C i-j when applied herein to a class of substituents, is meant to refer to embodiments of this invention for which each and every one of the number of carbon members, from i to j including i and j, is independently realized.
  • the term C 1-3 refers independently to embodiments that have one carbon member (C 1 ), embodiments that have two carbon members (C 2 ), and embodiments that have three carbon members (C 3 ).
  • C n-m alkyl refers to an aliphatic chain, whether straight or branched, with a total number N of carbon members in the chain that satisfies n ⁇ N ⁇ m, with m>n.
  • any disubstituent referred to herein is meant to encompass the various attachment possibilities when more than one of such possibilities are allowed.
  • X 4 is N and each of X 5 , X 6 , and X 7 is CR a , with R a independently chosen for X 5 , X 6 , and X 7 , where R a is H, methyl, chloro, or fluoro.
  • X 5 is N and each of X 4 , X 6 , and X 7 is CH.
  • each of X 4 , X 5 , and X 7 is CH and X 6 is N.
  • each of X 4 and X 7 is N and each of X 5 and X 6 is CH.
  • R a is H.
  • each of R 1 and R 2 is independently H, cyclopropyl, methyl, ethyl, propyl, hydroxyethyl, cyclopropylmethyl, benzyl, 1-phenylethyl, or 2-piperidin-1-yl-ethylamino.
  • R 1 and R 2 taken together with the nitrogen to which they are attached form pyrrolidine, piperidine, morpholine, piperazine, dihydroisoindole, tetrahydroquinoline, or tetrahydroisoquinoline, unsubstituted or substituted with one or two R d substituents.
  • each R d substituent is independently hydroxy, methyl, trifluoromethyl, hydroxymethyl, 1-hydroxy-1-methylethyl, fluoro, ethoxycarbonyl, carboxy, carbamoyl, phenyl, 3-trifluoromethylphenyl, 2-methoxyphenyl, 4-chlorophenyl, benzyl, pyridin-4-yl, pyridin-2-yl, pyrimidin-2-yloxy, pyridin-3-yloxy, phenoxy, phenylsulfanyl, 4-chlorophenylsulfanyl, pyridin-2-yloxy, pyridin-4-yloxy, or pyrrolidin-2-onyl.
  • R 1 and R 2 taken together with the nitrogen to which they are attached form 2,5-diaza-bicyclo[2.2.1]hept-2-yl, hexahydro-pyrrolo[3,4-c]pyrrol-2(1H)-yl, 3,8-diaza-bicyclo[3.2.1]oct-8-yl, or 3-amino-8-aza-bicyclo[3.2.1]oct-8-yl, each substituted with R e .
  • R e is acetyl or carbamoyl.
  • A is —CH 2 —. In other embodiments, A is —CH 2 CH 2 —. In still other embodiments, A is —OCH 2 CH 2 —.
  • chemical entities of the present invention are selected from the group consisting of:
  • chemical entities of the present invention are selected from the group consisting of:
  • chemical entities of the present invention are selected from the group consisting of:
  • chemical entities of the present invention are selected from the group consisting of:
  • chemical entities of the present invention are selected from the group consisting of:
  • the invention includes also pharmaceutically acceptable salts of the compounds represented by Formula (I), preferably of those described above and of the specific compounds exemplified herein, and methods using such salts.
  • a “pharmaceutically acceptable salt” is intended to mean a salt of a free acid or base of a compound represented by Formula (I) that is non-toxic, biologically tolerable, or otherwise biologically suitable for administration to the subject. See, generally, S. M. Berge, et al., “Pharmaceutical Salts”, J. Pharm. Sci., 1977, 66:1-19, and Handbook of Pharmaceutical Salts, Properties, Selection, and Use , Stahl and Wermuth, Eds., Wiley-VCH and VHCA, Zurich, 2002.
  • Preferred pharmaceutically acceptable salts are those that are pharmacologically effective and suitable for contact with the tissues of patients without undue toxicity, irritation, or allergic response.
  • a compound of Formula (I) may possess a sufficiently acidic group, a sufficiently basic group, or both types of functional groups, and accordingly react with a number of inorganic or organic bases, and inorganic and organic acids, to form a pharmaceutically acceptable salt.
  • pharmaceutically acceptable salts include sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, phosphates, monohydrogen-phosphates, dihydrogenphosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, propionates, decanoates, caprylates, acrylates, formates, isobutyrates, caproates, heptanoates, propiolates, oxalates, malonates, succinates, suberates, sebacates, fumarates, maleates, butyne-1,4-dioates, hexyne-1,6-dioates, benzoates,
  • the desired pharmaceutically acceptable salt may be prepared by any suitable method available in the art, for example, treatment of the free base with an inorganic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, sulfamic acid, nitric acid, boric acid, phosphoric acid, and the like, or with an organic acid, such as acetic acid, phenylacetic acid, propionic acid, stearic acid, lactic acid, ascorbic acid, maleic acid, hydroxymaleic acid, isethionic acid, succinic acid, valeric acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, oleic acid, palmitic acid, lauric acid, a pyranosidyl acid, such as glucuronic acid or galacturonic acid, an alpha-hydroxy acid, such as mandelic acid, citric acid, or tartaric acid, an inorganic acid, such as hydrochloric acid,
  • Embodiments of salts of this invention are prepared by adding the corresponding acid to the base form of compounds of this invention.
  • Illustrative examples of salts prepared with embodiments of this invention include acetates, formates, fumarates, citrates, hydrochlorides, tartrates, sulfates, phosphates, malates, malonates, bezoates and succinates.
  • Embodiments of salts of this invention were prepared by adding the corresponding acid to the base form of compounds of this invention. Some embodiments of salts according to this invention were characterized as being 1:1 as to the base/acid molar ratio.
  • the desired pharmaceutically acceptable salt may be prepared by any suitable method, for example, treatment of the free acid with an inorganic or organic base, such as an amine (primary, secondary or tertiary), an alkali metal hydroxide, alkaline earth metal hydroxide, any compatible mixture of bases such as those given as examples herein, and any other base and mixture thereof that are regarded as equivalents or acceptable substitutes in light of the ordinary level of skill in this technology.
  • an inorganic or organic base such as an amine (primary, secondary or tertiary), an alkali metal hydroxide, alkaline earth metal hydroxide, any compatible mixture of bases such as those given as examples herein, and any other base and mixture thereof that are regarded as equivalents or acceptable substitutes in light of the ordinary level of skill in this technology.
  • suitable salts include organic salts derived from amino acids, such as glycine and arginine, ammonia, carbonates, bicarbonates, primary, secondary, and tertiary amines, and cyclic amines, such as benzylamines, pyrrolidines, piperidine, morpholine, and piperazine, and inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum, and lithium.
  • amino acids such as glycine and arginine
  • ammonia carbonates, bicarbonates, primary, secondary, and tertiary amines
  • cyclic amines such as benzylamines, pyrrolidines, piperidine, morpholine, and piperazine
  • inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum, and lithium.
  • pharmaceutically acceptable salts of compounds of Formula (I) were hydrochloride, phosphate, sulfate, acetate, citrate, L-tartrate, or succinate salts.
  • compounds of Formula (I) were obtained as hydrochloride, phosphate, succinate or sulfate salts.
  • hydrochloride, or succinate salts of compounds of Formula (I) were obtained in crystalline form.
  • the invention also relates to pharmaceutically acceptable prodrugs of the compounds of Formula (I), and methods employing such pharmaceutically acceptable prodrugs.
  • prodrug means a precursor of a designated compound that, following administration to a subject, yields the compound in vivo via a chemical or physiological process such as solvolysis or enzymatic cleavage, or under physiological conditions (e.g., a prodrug on being brought to physiological pH is converted to the compound of Formula (I)).
  • a “pharmaceutically acceptable prodrug” is a prodrug that is non-toxic, biologically tolerable, and otherwise biologically suitable for administration to the subject. Illustrative procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in “Design of Prodrugs”, ed. H. Bundgaard, Elsevier, 1985.
  • prodrugs include compounds having an amino acid residue, or a polypeptide chain of two or more (e.g., two, three or four) amino acid residues, covalently joined through an amide or ester bond to a free amino, hydroxy, or carboxylic acid group of a compound of Formula (I).
  • amino acid residues include the twenty naturally occurring amino acids, commonly designated by three letter symbols, as well as 4-hydroxyproline, hydroxylysine, demosine, isodemosine, 3-methylhistidine, norvalin, beta-alanine, gamma-aminobutyric acid, citrulline homocysteine, homoserine, ornithine and methionine sulfone.
  • amides include those derived from ammonia, primary C 1-6 alkyl amines and secondary di(C 1-6 alkyl) amines. Secondary amines include 5- or 6-membered heterocycloalkyl or heteroaryl ring moieties. Examples of amides include those that are derived from ammonia, C 1-3 alkyl primary amines, and di(C 1-2 alkyl)amines.
  • esters of the invention include C 1-7 alkyl, C 5-7 cycloalkyl, phenyl, and phenyl(C 1-6 alkyl) esters.
  • Preferred esters include methyl esters.
  • Prodrugs may also be prepared by derivatizing free hydroxy groups using groups including hemisuccinates, phosphate esters, dimethylaminoacetates, and phosphoryloxymethyloxycarbonyls, following procedures such as those outlined in Adv. Drug Delivery Rev. 1996, 19, 115. Carbamate derivatives of hydroxy and amino groups may also yield prodrugs. Carbonate derivatives, sulfonate esters, and sulfate esters of hydroxy groups may also provide prodrugs.
  • acyloxy groups as (acyloxy)methyl and (acyloxy)ethyl ethers, wherein the acyl group may be an alkyl ester, optionally substituted with one or more ether, amine, or carboxylic acid functionalities, or where the acyl group is an amino acid ester as described above, is also useful to yield prodrugs.
  • Prodrugs of this type may be prepared as described in J. Med. Chem. 1996, 39, 10. Free amines can also be derivatized as amides, sulfonamides or phosphonamides. All of these prodrug moieties may incorporate groups including ether, amine, and carboxylic acid functionalities.
  • the present invention also relates to pharmaceutically active metabolites of compounds of Formula (I), and uses of such metabolites in the methods of the invention.
  • a “pharmaceutically active metabolite” means a pharmacologically active product of metabolism in the body of a compound of Formula (I) or salt thereof.
  • Prodrugs and active metabolites of a compound may be determined using routine techniques known or available in the art. See, e.g., Bertolini, et al., J. Med. Chem. 1997, 40, 2011-2016; Shan, et al., J. Pharm. Sci. 1997, 86 (7), 765-767; Bagshawe, Drug Dev. Res. 1995, 34, 220-230; Bodor, Adv. Drug Res.
  • the compounds of Formula (I) and their pharmaceutically acceptable salts, pharmaceutically acceptable prodrugs, and pharmaceutically active metabolites (collectively, “active agents”) of the present invention are useful as LTA4H modulators in the methods of the invention.
  • Such methods for modulating LTA4H activity comprise exposing LTA4H to an effective amount of at least one chemical entity selected from compounds of Formula (I), pharmaceutically acceptable salts of compounds of Formula (I), pharmaceutically acceptable prodrugs of compounds of Formula (I), and pharmaceutically active metabolites of compounds of Formula (I).
  • Embodiments of this invention inhibit LTA4H activity.
  • the LTA4H is in a subject with a disease, disorder, or medical condition mediated by LTA4H activity, such as those described herein. Symptoms or disease states are intended to be included within the scope of “medical conditions, disorders, or diseases.”
  • the invention relates to methods of using the active agents described herein to treat subjects diagnosed with or suffering from a disease, disorder, or condition mediated through LTA4H activity, such as inflammation.
  • Active agents according to the invention may therefore be used as an anti-inflammatory agents.
  • an active agent of the present invention is administered to treat inflammation.
  • Inflammation may be associated with various diseases, disorders, or conditions, such as inflammatory disorders, allergic disorders, dermatological disorders, autoimmune disease, lymphatic disorders, and immunodeficiency disorders, including the more specific conditions and diseases given below.
  • inflammatory diseases or inflammation-mediated diseases or conditions include, but are not limited to, acute inflammation, allergic inflammation, and chronic inflammation.
  • Illustrative types of inflammation treatable with an LTA4H modulating agent include inflammation due to any one of a plurality of conditions such as allergy, abdominal aortic aneurysm, asthma, nasal polyps, allergic rhinitis, nasal itch, ocular inflammation (e.g., post-surgical ocular inflammation), conjunctivitis, uveitis, dry eye, psoriasis, pruritis, itch, itchy skin, atopic dermatitis, urticaria (hives), contact dermatitis, scleroderma, skin burns, acne, inflammatory bowel diseases (including colitis, Crohn's disease and ulcerative colitis), chronic obstructed pulmonary disease (COPD), atherosclerosis, arthritis (including rheumatoid arthritis), multiple sclerosis, myocardial infarction, stroke, pain, gingivitis, bronchitis, cystic fibrosis, upper gastrointestinal cancer, sepsis, autoimmune thyroid diseases,
  • autoimmune diseases that lead to inflammation include Myasthenia gravis, autoimmune neuropathies, such as Guillain-Barré, autoimmune uveitis, autoimmune hemolytic anemia, pernicious anemia, autoimmune thrombocytopenia, temporal arteritis, anti-phospholipid syndrome, vasculitides, such as Wegener's granulomatosis, Behcet's disease, dermatitis herpetiformis, pemphigus vulgaris, vitiligio, primary biliary cirrhosis, autoimmune hepatitis, autoimmune oophoritis and orchitis, autoimmune disease of the adrenal gland, polymyositis, dermatomyositis, spondyloarthropathies, such as ankylosing spondylitis, Sjogren syndrome, and Sjogren-Larsson syndrome.
  • autoimmune neuropathies such as Guillain-Barré, autoimmune uveitis, autoimmune hemolytic anemia, per
  • Pruritis treatable with an LTA4H-modulating agent according to the invention includes that which is a symptom of allergic cutaneous diseases (such as atopic dermatitis and hives) and other metabolic disorders (such as chronic renal failure, hepatic cholestasis, and diabetes mellitus).
  • allergic cutaneous diseases such as atopic dermatitis and hives
  • other metabolic disorders such as chronic renal failure, hepatic cholestasis, and diabetes mellitus.
  • an active agent of the present invention is administered to treat allergy, aortic aneurysm, asthma, autoimmune diseases, pruritis, inflammatory bowel disease, ulcerative colitis, or cardiovascular disease, including atherosclerosis and prevention of myocardial infarction.
  • an active agent of the present invention alone or in combination with some other agent, is administered to treat aortic aneurysms, delaying the time to or avoiding the surgical intervention to repair aortic aneurysms, slowing the progression of aortic aneurysms, or avoiding or slowing down the progression towards or the incidence of aortic rupture.
  • an active agent alone or in combination with some other agent, is administered for any of such treatments when the aortic aneurysm is an abdominal aortic aneurysm.
  • examples of embodiments of such other agent are given by CysLT receptor antagonists and LTC4 synthase inhibitors.
  • the active agents may be used to treat subjects diagnosed with or suffering from a disease, disorder, or condition mediated through LTA4H activity.
  • the term “treat” or “treating” as used herein is intended to refer to administration of an active agent or composition of the invention to a subject for the purpose of effecting a therapeutic or prophylactic benefit through modulation of LTA4H activity. Treating includes reversing, ameliorating, alleviating, inhibiting the progress of, lessening the severity of, or preventing a disease, disorder, or condition, or one or more symptoms of such disease, disorder or condition mediated through modulation of LTA4H activity.
  • subject refers to a mammalian patient in need of such treatment, such as a human.
  • Modules include both inhibitors and activators, where “inhibitors” refer to compounds that decrease, prevent, inactivate, desensitize or down-regulate LTA4H expression or activity, and “activators” are compounds that increase, activate, facilitate, sensitize, or up-regulate LTA4H expression or activity.
  • embodiments of chemical entities according to this invention are LTA4H-modulating chemical entities.
  • an effective amount of at least one active agent according to the invention is administered to a subject suffering from or diagnosed as having such a disease, disorder, or condition.
  • An “effective amount” means an amount or dose sufficient to generally bring about the desired therapeutic or prophylactic benefit in patients in need of such treatment for the designated disease, disorder, or condition.
  • an “effective amount” means an amount sufficient to at least affect the activity of such receptor. Measuring the activity of the target receptor may be performed by routine analytical methods. Target receptor modulation is useful in a variety of settings, including assays and treating conditions modulated through LTA4H activity.
  • effective amounts or doses of the active agents of the present invention may be ascertained by routine methods such as modeling, dose escalation studies or clinical trials, and by taking into consideration routine factors, e.g., the mode or route of administration or drug delivery, the pharmacokinetics of the agent, the severity and course of the disease, disorder, or condition, the subject's previous or ongoing therapy, the subject's health status and response to drugs, and the judgment of the treating physician.
  • routine methods such as modeling, dose escalation studies or clinical trials, and by taking into consideration routine factors, e.g., the mode or route of administration or drug delivery, the pharmacokinetics of the agent, the severity and course of the disease, disorder, or condition, the subject's previous or ongoing therapy, the subject's health status and response to drugs, and the judgment of the treating physician.
  • An exemplary dose is in the range of from about 0.001 to about 200 mg of active agent per kg of subject's body weight per day, preferably from about 0.05 to about 100 mg/kg/day, or from about 0.5 to about 35 mg/kg/day, or from about 0.5 to about 20 mg/kg/day, or from about 0.1 to about 10 mg/kg daily in single or divided dosage units (e.g., BID, TID, QID).
  • a suitable dosage amount is from about 0.02 to about 7 g/day, or from about 0.2 to about 2.5 g/day.
  • Dosages from about 20 mg/day to about 60 mg/day are contemplated. In some embodiments, such dosages would be administered once daily.
  • Examples of embodiments of this invention are given by tablets containing from about 0.005 mol free base per tablet to about 0.5 mol free base per tablet. Other embodiments are given by tablets containing from about 0.005 mol free base per tablet to about 0.01 mol free base per tablet. Additional embodiments are given by tablets containing from about 0.03 mol free base per tablet to about 0.06 mol free base per tablet. Further embodiments are given by tablets containing from about 0.3 mol free base per tablet to about 0.6 mol free base per tablet. Some embodiments of this invention were prepared with about 0.0095 mol free base per tablet. Other embodiments of this invention were prepared with about 0.047 mol free base per tablet. Still other embodiments of this invention were prepared with about 0.47 mol free base per tablet.
  • the dose may be adjusted for preventative or maintenance treatment.
  • the dosage or the frequency of administration, or both may be reduced as a function of the symptoms, to a level at which the desired therapeutic or prophylactic effect is maintained.
  • treatment may cease. Patients may, however, require intermittent treatment on a long-term basis upon any recurrence of symptoms.
  • the active agents of the invention may be used in combination with additional active ingredients in the treatment of the above conditions or with other active ingredients.
  • Montelukast salts are examples of such additional active ingredients, such as montelukast sodium.
  • Conditions that are mediated by LTA4H activity, such as asthma for example, could be treated by embodiments of this invention such as active agents of this invention alone or in combination with others, such as montelukast salts.
  • the additional active ingredients may be coadministered separately with an active agent of Formula (I) or included with such an agent in a pharmaceutical composition according to the invention.
  • additional active ingredients are those that are known or discovered to be effective in the treatment of conditions, disorders, or diseases mediated by LTA4H activity, such as another LTA4H modulator or a compound active against another target associated with the particular condition, disorder, or disease.
  • the combination may serve to increase efficacy (e.g., by including in the combination a compound potentiating the potency or effectiveness of an agent according to the invention), decrease one or more side effects, or decrease the required dose of the active agent according to the invention.
  • inventions of this invention further comprise the administration of at least one CysLT receptor antagonist (for example, Montelukast/Singulair®) and/or at least one LTC4 synthase inhibitor.
  • CysLT receptor antagonists are CysLT1 and CysLT2 antagonists.
  • compositions of the invention comprise an effective amount of at least one active agent in accordance with the invention.
  • Such compositions may further comprise a pharmaceutically acceptable excipient.
  • a “pharmaceutically acceptable excipient” refers to a substance that is non-toxic, biologically tolerable, and otherwise biologically suitable for administration to a subject, such as an inert substance, added to a pharmacological composition or otherwise used as a vehicle, carrier, or diluent to facilitate administration of a agent and that is compatible therewith.
  • excipients include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils, and polyethylene glycols.
  • Suitable excipients may also include antioxidants. Such antioxidants may be used in a pharmaceutical composition or in a storage medium to prolong the shelf-life of the drug product.
  • compositions containing one or more dosage units of the active agents may be prepared using suitable pharmaceutical excipients and compounding techniques known or that become available to those skilled in the art.
  • the compositions may be administered in the inventive methods by a suitable route of delivery, e.g., oral, parenteral, rectal, topical, or ocular routes, or by inhalation.
  • the preparation may be in the form of tablets, capsules, sachets, dragees, powders, granules, lozenges, powders for reconstitution, liquid preparations, or suppositories.
  • the compositions are formulated for intravenous infusion, topical administration, or oral administration.
  • compounds of the present invention are orally active inhibitors of LTA4H.
  • Some embodiments of this invention were prepared with a round tablet image, other embodiments were prepared with a capsule-shaped tablet image, and still other embodiments were prepared with an oval tablet image. Further embodiments of tablet images were prepared with masses of about 100 mg, 500 mg and 1000 mg.
  • the active agents of the invention can be provided in the form of tablets or capsules, or as a solution, emulsion, or suspension.
  • the active agents may be formulated to yield a dosage of, e.g., from about 0.05 to about 50 mg/kg daily, or from about 0.05 to about 20 mg/kg daily, or from about 0.1 to about 10 mg/kg daily, or from about 0.2 to about 1 mg/kg daily.
  • Oral tablets may include the active ingredient(s) mixed with compatible pharmaceutically acceptable excipients such as diluents, disintegrating agents, binding agents, lubricating agents, sweetening agents, flavoring agents, coloring agents and preservative agents.
  • suitable inert fillers include sodium and calcium carbonate, sodium and calcium phosphate, lactose, starch, sugar, glucose, methyl cellulose, magnesium stearate, mannitol, sorbitol, and the like.
  • Exemplary liquid oral excipients include ethanol, glycerol, water, and the like.
  • Starch, polyvinyl-pyrrolidone (PVP), sodium starch glycolate, microcrystalline cellulose, and alginic acid are exemplary disintegrating agents.
  • Binding agents may include starch and gelatin.
  • the lubricating agent if present, may be magnesium stearate, stearic acid or talc. If desired, the tablets may be coated with a material such as glyceryl monostearate or glyceryl distearate to delay absorption in the gastrointestinal tract, or may be coated with an enteric coating.
  • Lactose Fast Flo #316 is one example of a filler that was used in the preparation of embodiments of this invention.
  • Avicel PH102 is another example of a filler that was used in the preparation of embodiments of this invention.
  • Polyplasdone XL-10 is an example of a disintegrant that was used in the preparation of embodiments of this invention.
  • Magnesium stearate is an example of a lubricant that was used in the preparation of embodiments of this invention.
  • Yellow ferroxide is an example of a pigment that was used in the preparation of embodiments of this invention.
  • Intragranular excipients that were used in the preparation of embodiments of this invention are illustratively given by fillers, disintegrants, lubricants and pigments, such as lactose fast flow # 316, avicel PH102, polyplasdone XL-10, yellow ferroxide and magnesium stearate.
  • Extragranular excipients that were used in the preparation of embodiments of this invention are illustratively given by lubricants and disintegrants, such as magnesium stearate and polyplasdone XL-10.
  • Capsules for oral administration include hard and soft gelatin capsules.
  • active ingredient(s) may be mixed with a solid, semi-solid, or liquid diluent.
  • Soft gelatin capsules may be prepared by mixing the active ingredient with water, an oil such as peanut oil or olive oil, liquid paraffin, a mixture of mono and di-glycerides of short chain fatty acids, polyethylene glycol 400, or propylene glycol.
  • Embodiments of this invention are provided by tablet forms with an amount of active compound corresponding to from about 1% to about 30% of free base.
  • Other embodiments contain active compound corresponding to from about 5% to about 25% of free base.
  • Still other embodiments contain active compound corresponding to from about 1% to about 10% of free base.
  • Further embodiments contain active compound corresponding to from about 20% to about 30% of free base.
  • Additional embodiments contain active compound corresponding to from about 10% to about 20% of free base.
  • Embodiments of this invention in the form of tablet doses of about 5 mg per tablet and about 25 mg per tablet were prepared in the form of compositions with active compound corresponding to about 5% free base.
  • Embodiments of this invention in the form of tablet doses of about 250 mg per tablet were prepared in the form of compositions with active compound corresponding to about 25% free base. Actual amounts depended on the salt of choice.
  • lactose can be used as the adjustable excipient for suitable batch correction depending on the specific salt form being used.
  • avicel can be used as the adjustable excipient for suitable batch correction depending on the specific salt form being used.
  • Embodiments of this invention in the form of tablets comprise from about 70% to about 95% of non-active intragranular excipients and from about 0.2% to about 4% of non-active extragranular excipients.
  • Embodiments of this invention in the form of tablets comprise from about 90% to about 95% of non-active intragranular excipients and from about 0.2% to about 0.3% of non-active extragranular excipients.
  • Embodiments of this invention in the form of tablets comprise from about 70% to about 75% of non-active intragranular excipients and from about 3% to about 4% of non-active extragranular excipients.
  • Embodiments of this invention in the form of tablets comprise from about 65% to about 95% of intragranular filler.
  • Embodiments of this invention in the form of tablets comprise from about 90% to about 95% of intragranular filler.
  • Embodiments of this invention in the form of tablets comprise from about 65% to about 70% of intragranular filler.
  • Embodiments of this invention in the form of tablets comprise from about 2.5% to about 3.5% of intragranular disintegrant.
  • Embodiments of this invention in the form of tablets comprise from about 0.2% to about 0.4% of intragranular pigment.
  • Embodiments of this invention in the form of tablets comprise from about 0.1% to about 1.0% of intragranular lubricant.
  • Embodiments of this invention in the form of tablets comprise from about 0.1% to about 1.0% of extragranular lubricant.
  • Embodiments of this invention in the form of tablets comprise from about 2.5% to about 3.5% of extragranular disintegrant.
  • Embodiments of this invention in the form of tablets comprised about 91.2% intragranular filler, about 3.0% intragranular disintegrant, about 0.30% intragranular pigment, and about 0.25% intragranular lubricant.
  • Embodiments of this invention in the form of tablets comprised about 0.25% extragranular lubricant.
  • Embodiments of this invention in the form of tablets comprised about 67.7% intragranular filler, about 3.0% intragranular disintegrant, about 0.30% intragranular pigment, and about 0.75% intragranular lubricant.
  • Embodiments of this invention in the form of tablets comprised about 0.25% extragranular lubricant and about 3.0% extragranular disintegrant.
  • Tabletting equipment used in the preparation of some embodiments of this invention comprised standard technology used to this effect, including 60 mesh sieve and balance for weighing, TFC labo roller compactor for compaction, TFC Labo granulator for granulating, Bohle bin blender for blending, and Piccola press for tableting with various punch sets depending on the choice of tablet image.
  • Tabletting process used in embodiments of this invention comprised low shear blending of excipients, low shear blending with lubricant, milling and tablet formation. Some process embodiments included geometric blending.
  • Liquids for oral administration may be in the form of suspensions, solutions, emulsions or syrups or may be lyophilized or presented as a dry product for reconstitution with water or other suitable vehicle before use.
  • Such liquid compositions may optionally contain: pharmaceutically-acceptable excipients such as suspending agents (for example, sorbitol, methyl cellulose, sodium alginate, gelatin, hydroxyethylcellulose, carboxymethylcellulose, aluminum stearate gel and the like); non-aqueous vehicles, e.g., oil (for example, almond oil or fractionated coconut oil), propylene glycol, ethyl alcohol, or water; preservatives (for example, methyl or propyl p-hydroxybenzoate or sorbic acid); wetting agents such as lecithin; and, if desired, flavoring or coloring agents.
  • suspending agents for example, sorbitol, methyl cellulose, sodium alginate, gelatin, hydroxyethylcellulose, carboxymethylcellulose,
  • compositions may be formulated for rectal administration as a suppository.
  • parenteral use including intravenous, intramuscular, intraperitoneal, or subcutaneous routes, the agents of the invention may be provided in sterile aqueous solutions or suspensions, buffered to an appropriate pH and isotonicity or in parenterally acceptable oil.
  • Suitable aqueous vehicles include Ringer's solution and isotonic sodium chloride.
  • Such forms may be presented in unit-dose form such as ampules or disposable injection devices, in multi-dose forms such as vials from which the appropriate dose may be withdrawn, or in a solid form or pre-concentrate that can be used to prepare an injectable formulation.
  • Illustrative infusion doses range from about 1 to 1000 ⁇ g/kg/minute of agent admixed with a pharmaceutical carrier over a period ranging from several minutes to several days.
  • Active agents may alternatively be administered in methods of this invention by inhalation, via the nasal or oral routes, e.g., in a spray formulation also containing a suitable carrier.
  • compounds B1 are converted to their corresponding sodium or potassium salts before reaction with compounds A3 in a solvent such as DMF, tetrahydrofuran (THF) or diethyl ether.
  • a solvent such as DMF, tetrahydrofuran (THF) or diethyl ether.
  • R 10 is -A-NR 1 R 2
  • compounds B2 are compounds of Formula (I).
  • Compounds B2 where R 10 is -A-OH are then converted into compounds B2 where R 10 is —CHO by oxidation methods known in the art.
  • compounds B2 where R 10 is an ester may be reduced to the corresponding aldehydes where R 10 is —CHO or —CH 2 CHO.
  • Such aldehydes are optionally converted to the corresponding bisulfite adducts for purification and/or storage.
  • compounds B2 where R 10 is -A-OH are converted into compounds B2 where R 10 is -A-LG by standard halogenation or sulfonylation methods known in the art.
  • such alcohols are reacted with methanesulfonic anhydride and a tertiary amine base in a solvent such as dichloromethane or dichloroethane, or with thionyl chloride, oxalyl chloride, or POCl 3 , neat or in a solvent such as dichloromethane, 1,2-dichloroethane (DCE), toluene, or acetonitrile.
  • amines HNR 1 R 2 are used in free base form, and in some embodiments, amines HNR 1 R 2 are used in the corresponding salt form (such as a hydrochloride salt form).
  • salt form such as a hydrochloride salt form.
  • One skilled in the art will recognize amines HNR 1 R 2 containing an additional amine functionality may have such functionality optionally protected with a suitable nitrogen protecting group.
  • Reactions are preferably performed in the presence of a suitable base (such as Et 3 N, iPr 2 NEt, pyridine, K 2 CO 3 , Cs 2 CO 3 , Na 2 CO 3 , NaHCO 3 , or K 3 PO 4 ), in a polar solvent (such as acetonitrile, DMF, MeOH, EtOH, isopropanol, tert-butanol, or tert-amyl alcohol) to give compounds of Formula (I).
  • a suitable base such as Et 3 N, iPr 2 NEt, pyridine, K 2 CO 3 , Cs 2 CO 3 , Na 2 CO 3 , NaHCO 3 , or K 3 PO 4
  • a polar solvent such as acetonitrile, DMF, MeOH, EtOH, isopropanol, tert-butanol, or tert-amyl alcohol
  • compounds of Formula (I) are prepared from compounds B2, where R 10 is —CHO or —CH 2 CHO.
  • Aldehydes B2 may optionally be used or purified in a protected form, such as a bisulfite complex.
  • Reaction of aldehydes B2 with amines HNR 1 R 2 (used as free amines or corresponding salts) in the presence of a suitable reducing agent (such as NaCNBH 3 or NaB(OAc) 3 H) in a solvent such as DCE, CH 2 Cl 2 , MeOH, or EtOH, and optionally employing an acid catalyst (such as acetic acid or ZnCl 2 ), provides compounds of Formula (I).
  • the reaction with the corresponding HNR 1 R 2 salts may optionally employ a base, such as Et 3 N, to produce the free amine in situ.
  • compounds of Formula (I) are prepared as shown in Scheme D.
  • Compounds D1 are reacted with phenols D2 to give compounds D3.
  • reactions are performed in the presence of a suitable base, such as K 2 CO 3 , Cs 2 CO 3 , Na 2 CO 3 , NaHCO 3 , or K 3 PO 4 , in a polar solvent such as DMF, acetonitrile, MeOH, EtOH, isopropanol, or tert-butanol.
  • a suitable base such as K 2 CO 3 , Cs 2 CO 3 , Na 2 CO 3 , NaHCO 3 , or K 3 PO 4
  • a polar solvent such as DMF, acetonitrile, MeOH, EtOH, isopropanol, or tert-butanol.
  • reactions are performed using K 2 CO 3 or Cs 2 CO 3 , in DMF or acetonitrile.
  • compounds D3 are converted to chlorides D4 by chlorination methods.
  • reactions are accomplished by treatment with thionyl chloride, oxalyl chloride, or POCl 3 , neat or in a solvent such as dichloromethane, dichloroethane, toluene, or acetonitrile.
  • reactions are accomplished by reaction with thionyl chloride in dichloromethane.
  • Compounds D4 are then reacted with amines HNR 1 R 2 to give compounds of Formula (I).
  • reactions are performed in the presence of a suitable base, such as Et 3 N, pyridine, K 2 CO 3 , Cs 2 CO 3 , Na 2 CO 3 , NaHCO 3 , or K 3 PO 4 , in a polar solvent, such as acetonitrile, DMF, MeOH, EtOH, isopropanol, or tert-butanol.
  • a suitable base such as Et 3 N, pyridine, K 2 CO 3 , Cs 2 CO 3 , Na 2 CO 3 , NaHCO 3 , or K 3 PO 4
  • a polar solvent such as acetonitrile, DMF, MeOH, EtOH, isopropanol, or tert-butanol.
  • reactions are performed using K 2 CO 3 or Cs 2 CO 3 in acetonitrile.
  • Compound BX is reacted in the presence of a suitable reducing agent, such as NaB(OAc) 3 H, NaCNBH 3 , and chemically compatible mixtures thereof, in a solvent, such as MeOH, EtOH, 2-propanol, acetonitrile, DCE, CH 2 Cl 2 , and chemically compatible mixtures thereof, optionally in the presence of an acid catalyst, such as acetic acid, with compound HNR 1 R 2 to make F1, where HNR 1 R 2 is used in the free amine form, in a salt form or in the form of mixtures thereof.
  • a suitable reducing agent such as NaB(OAc) 3 H, NaCNBH 3 , and chemically compatible mixtures thereof
  • a solvent such as MeOH, EtOH, 2-propanol, acetonitrile, DCE, CH 2 Cl 2
  • an acid catalyst such as acetic acid
  • R x is one of —CHO and —CH 2 CHO
  • a X is one of —CH 2 — and —CH 2 CH 2 —.
  • Compound F1 is reacted with A3 in the presence of a suitable base, such as K 2 CO 3 , Cs 2 CO 3 , Na 2 CO 3 , NaHCO 3 , K 3 PO 4 , and chemically compatible mixtures thereof, in a polar solvent such as DMF, acetonitrile, methanol (MeOH), ethanol (EtOH), isopropanol, tert-butanol, and chemically compatible mixtures thereof, to form compound (I x ),
  • a suitable base such as K 2 CO 3 , Cs 2 CO 3 , Na 2 CO 3 , NaHCO 3 , K 3 PO 4 , and chemically compatible mixtures thereof
  • a polar solvent such as DMF, acetonitrile, methanol (MeOH), ethanol (EtOH), isopropano
  • Synthesis according to Scheme F avoids the use of halogenated solvents and thionyl chloride, avoids the formation of halo-substituted intermediates, is shorter than other synthetic processes, and reduces the risk of side-reactions and byproducts.
  • R 1 i are non-acid sensitive protecting groups and R 1 i are acylated functional groups derived via reacting with substituted acid anhydrides.
  • R 1 i is one of H, C 1-6 alkylC(O)—, arylC(O)—, and EstOC(O)—, wherein the moiety “Est” signifies that the carboxy group is in some embodiments in an ester form.
  • the moieties C 1-6 alkyl (linear or branched) and aryl in R 1 i are optionally substituted with at least one substituent such as halo and linear or branched C 1-6 alkyl.
  • R 2 i is one of H, C 1-10 alkyl (linear or branched), —CH 2 aryl, —S(O) 2 aryl, and —S(O) 2 C 1-6 alkyl.
  • the moieties linear or branched C 1-10 alkyl and aryl in R 2 i are optionally substituted with at least one substituent such as halo and C 1-6 alkyl.
  • the synthesis is a two-step process involving the reduction of substituted bicyclic oximes to primary amines via hydrogenation, transfer hydrogenation, aluminium-nickel alloy or Na metal then acetylation to form the bicyclic acetamide. It is known that in acidic or basic hydrogenation conditions, certain bicyclic endo stereochemistry can be favored over exo but frequently as mixtures of both. It was found in the context of this invention that the implementation of conventional synthetic conditions leads to poor endo selectivity and/or mixtures of the endo form with other species such as the following ketones and/or dimers:
  • compounds E5 1 are prepared according to Scheme E1 by acetylation and reduction of oxime E4 1 .
  • E5 1 is desired in a final form as a secondary amine (R 2 i then being H)
  • suitable protection of this group can then be implemented, such as by choosing another acceptable form of substituent R 2 i as described herein.
  • R 2 i′ is one of the R 2 i substituents except for H. Therefore, some embodiments of Formula (I) are available according to Schemes A-D using compound E5 1 as the amine HNR 1 R 2 .
  • Compounds E5 1 are obtained from the reduction of oxime E4 1 , which is commercially available or can be conventionally obtained from tropinone. Embodiments of synthetic methodologies according to this invention generate compounds E5 1 from compounds E4 1 in a single reaction step, with high selectivity, and with simplified isolation procedures. In some embodiments, compounds E4 1 are reacted with carboxylic acid anhydrides and hydrogen in the presence of a suitable hydrogenation catalyst in a chemically compatible solvent.
  • the intermediate HNR 1 R 2 is compound N-[(3-endo)-8-azabicyclo[3.2.1]oct-3-yl]acetamide (E6).
  • compound E6 is prepared according to Scheme E. Therefore, some embodiments of Formula (I) are available according to Schemes A-D using compound E6 as the amine HNR 1 R 2 .
  • Compound E6 is prepared by debenzylation of compound E5.
  • Compound E5 is available by reduction of oxime compound E4, which is commercially available (See also, U.S. Pat. No. 4,432,983), and generally prepared from tropinone (E1) as described in the art.
  • Compounds E5 and E6 are known (See, Eur. J. Med. Chem.
  • the desired functionalized amine is prepared in a single step with high endo selectivity over the corresponding enamine:
  • the compound E4 is reacted with hydrogen in the presence of acetic anhydride to form compound E5 in a single step.
  • reactions are performed by reacting a compound E4 with carboxylic acid anhydrides (such as acetic, propionic, ethylbutyric, butyric, isobutyric, valeric, isovaleric, trimethyl acetic, and trifluoroacetic anhydrides) and hydrogen in the presence of a suitable catalyst (such as Pt/C, Raney Ni, Rh/C, or a mixture thereof), in a solvent such as ethyl acetate, acetic acid, MeOH, EtOH, isopropanol, or a mixture thereof.
  • carboxylic acid anhydrides such as acetic, propionic, ethylbutyric, butyric, isobutyric, valeric, isovaleric, trimethyl acetic, and trifluoroacetic anhydrides
  • a suitable catalyst such as Pt/C
  • the reaction of a compound E4 with acetic anhydride and hydrogen is performed in ethyl acetate, with Pt/C as the catalyst, and with added acetic acid.
  • a continuous flow hydrogenation H-Cube MidiTM instrument was used with Pt/C catalyst to react compound E4 with ethyl acetate, acetic anhydride and acetic acid, a reaction that provided endo E5.
  • the ethyl acetate, acetic anhydride and acetic acid mixture concentration to obtain an endo/enamine selectivity of about 95/5 was approximately 0.1M.
  • Methods of the invention further comprise reacting a compound E5 to form N-[(3-endo)-8-azabicyclo[3.2.1]oct-3-yl]acetamide (compound E6) as described herein.
  • Embodiments of this invention included reactions performed with 10% Pt/C catalyst amounts ranging from about 0.015 g to about 0.15 g, with about 10 eq acetic anhydride in EtOAc-based solvents in the presence of various amounts of AcOH.
  • Other embodiments included reactions performed with 5% Pt/C catalyst amounts ranging from about 0.025 g to about 0.25 g, with about 10 eq to about 40 eq acetic anhydride in EtOAc-based solvents in the presence of various amounts of AcOH.
  • Still other embodiments included reactions performed with Raney Ni catalyst, with about 2 eq to about 10 eq acetic anhydride in solvents such as EtOAc, acetic acid, and EtOAc-based solvents in the presence of various amounts of AcOH.
  • Embodiments of this invention that included the use of a continuous flow hydrogenation had a variety of reaction conditions, such as ethyl acetate, acetic anhydride and acetic acid mixture concentrations ranging from about 0.01M to about 0.6M, use of 10% Pt/C calyst, solution flow rates of about 3 ml/min, hydrogen flow rates of about 45 ml/min, at pressures of about 80 bar and temperatures of about 60° C.
  • the protecting group is a Boc group
  • deprotection is accomplished using an acid such as HCl or trifluoroacetic acid (TFA), in a solvent such as diethyl ether, dioxane, or CH 2 Cl 2 .
  • a solvent such as diethyl ether, dioxane, or CH 2 Cl 2 .
  • Additional substituents on the —NR 1 R 2 group are then installed by acylation or carbamoylation protocols using methods known in the art.
  • Compounds of Formula (I) may be converted to their corresponding salts using methods described in the art.
  • an amine of Formula (I) is treated with TFA, HCl, or citric acid in a solvent such as diethyl ether, CH 2 Cl 2 , THF, MeOH, or isopropanol to provide the corresponding salt form.
  • Compounds prepared according to the schemes described above may be obtained as single enantiomers, diastereomers, or regioisomers, by enantio-, diastero-, or regiospecific synthesis, or by resolution.
  • Compounds prepared according to the schemes above may alternately be obtained as racemic (1:1) or non-racemic (not 1:1) mixtures or as mixtures of diastereomers or regioisomers.
  • single enantiomers may be isolated using conventional separation methods known to one skilled in the art, such as chiral chromatography, recrystallization, diastereomeric salt formation, derivatization into diastereomeric adducts, biotransformation, or enzymatic transformation.
  • separation methods known to one skilled in the art, such as chiral chromatography, recrystallization, diastereomeric salt formation, derivatization into diastereomeric adducts, biotransformation, or enzymatic transformation.
  • regioisomeric or diastereomeric mixtures are obtained, single isomers may be separated using conventional methods such as chromatography or crystallization.
  • reaction mixtures were magnetically stirred at room temperature (rt). Where solutions were “dried,” they were generally dried over a drying agent such as Na 2 SO 4 or MgSO 4 . Where mixtures, solutions, and extracts were “concentrated”, they were typically concentrated on a rotary evaporator under reduced pressure. Column chromatography was typically conducted on disposable silica gel columns for flash chromatography (Teledyne Isco, Inc.). Microwave reactions were performed on a CEM Discover microwave reactor.
  • Analytical reversed phase LC/MS was performed either on an Agilent 1100 Series instrument using 5 to 99% acetonitrile/water (0.05% trifluoroacetic acid) over 5.0 min or 7.0 min with a flow rate of 0.6 mL/min (Waters XTerra RP18 (5 ⁇ m, 3.0 ⁇ 100 mm) column) or on a Waters 2790 instrument using 5 to 99% acetonitrile/water (0.1% formic acid) over 5.0 min with a flow rate of 0.6 mL/min (Waters XTerra RP18 (5 ⁇ m, 3.0 ⁇ 100 mm) column).
  • Preparative reversed phase HPLC was performed on a Dionex APS2000 LC/MS or HPLC with a Phenomenex Gemini C18 (5 ⁇ m, 30 ⁇ 100 mm) column or a Waters XBridge C18 (5 ⁇ m, 30 ⁇ 100 mm) column and variable gradients of acetonitrile/water (20 mM NH 4 OH) at a flow rate of 30 mL/min.
  • the purification was performed with a Phenomenex Gemini C18 (5 ⁇ m, 50 ⁇ 100 mm) column or a Waters XBridge C18 (5 ⁇ m, 50 ⁇ 100 mm) column and variable gradients of acetonitrile/water (20 mM NH 4 OH) at a flow rate of 80 mL/min.
  • Formate salts of desired compounds were obtained when purifications were performed using an Inertsil ODS-3 C18 (3 ⁇ m, 30 ⁇ 100 mm) column at 46° C. with variable gradients of acetonitrile/water (0.1% formic acid) at a flow rate of 90 mL/min.
  • Mass spectra were obtained on an Agilent series 1100 MSD using electrospray ionization (ESI) in positive mode unless otherwise indicated. Calculated (calcd.) mass corresponds to the exact mass.
  • Nuclear magnetic resonance (NMR) spectra were obtained on Bruker model DRX spectrometers.
  • the format of the 1 H NMR data below is: chemical shift in ppm downfield of the tetramethylsilane reference (multiplicity, coupling constant J in Hz, integration).
  • NMR interpretation was performed using MestReC or MestReNova software to assign chemical shift and multiplicity. In cases where two adjacent peaks of equal or unequal height were observed, these two peaks may be labeled either as a multiplet or as a doublet. In the case of a doublet a coupling constant using this software may be assigned. In any given example, one or more protons may not be reported due to obscurity by water and/or solvent peaks.
  • Chemical names were typically generated using ACD/Name Version 9 (Advanced Chemistry Development, Toronto, Ontario, Canada).
  • Method A Water was added to the reaction mixture, and the product was extracted with CH 2 Cl 2 and washed with 1 M NaOH. Charcoal was added to the organic layer, and the mixture stirred for 2 h. After drying and filtration through diatomaceous earth, the solution was concentrated to afford the desired product.
  • Method B The solids were removed by filtration and washed with CH 3 CN (100 mL). To the filtrate was added an aqueous solution of NaHSO 3 (22 g, 147 mmol, 54 mL water). After stirring for 3.5 h, the mixture was filtered and the wet cake was dried under vacuum overnight to afford the bisulfite complex as a white powder (57.2 g). To a solution of this bisulfite complex (57.2 g) in CH 2 Cl 2 (520 mL) was added an aqueous solution of NaOH (8 g in 540 mL water, 1.25 equiv.). The resulting mixture was vigorously stirred at rt for 2 h.
  • Step 1 meso-8-Azabicyclo[3.2.1]octan-3-one hydrochloride.
  • a solution of tropinone (1.0 equiv.) in toluene (1.2 M) was treated with 1-chloroethyl chloroformate (1.5 equiv.).
  • the reaction mixture was heated at reflux overnight (18 h) then cooled to rt and concentrated to a brown oil.
  • MeOH 1.2 M relative to starting material
  • Step 2 meso-8-Benzyl-8-azabicyclo[3.2.1]octan-3-one.
  • a mixture of meso-8-azabicyclo[3.2.1]octan-3-one hydrochloride (1.0 equiv.), benzyl bromide (1.0 equiv.), and Na 2 CO 3 (2.5 equiv.) in CH 3 CN (0.8 M) was heated at reflux for 2 h and then concentrated to half the original volume. The mixture was quenched with water (1.2 M relative to starting material) and tert-butyl methyl ether (1.2 M relative to starting material) and then, with vigorous stirring, slowly acidified to pH 1-2 with concentrated HCl.
  • Step 3 meso-8-Benzyl-8-azabicyclo[3.2.1]octan-3-one oxime.
  • Hydroxylamine hydrochloride (2.0 equiv.) was added causing mild exotherm.
  • NaHCO 3 was added in six portions over a period of 15 min to minimize gas evolution.
  • the reaction mixture was heated to 50° C. for 1 h, becoming cloudy in appearance before precipitation occurred.
  • Step 4 meso-N-[(3-endo)-8-Benzyl-8-azabicyclo[3.2.1]oct-3-yl]acetamide.
  • Step 5 meso-N-[(3-endo)-8-Azabicyclo[3.2.1]oct-3-yl]acetamide.
  • EtOH 0.5 M
  • Pd(OH) 2 16% wt
  • the mixture was agitated under 55 psi H 2(g) overnight at rt.
  • the catalyst was filtered and washed with EtOH (1.2 M relative to starting material). The filtrate was concentrated to a white solid then dried overnight to afford the title compound (100%).
  • This intermediate (1.22 g, 4.72 mmol) was dissolved in ethanol (12 mL) and added onto a mixture of 20% palladium hydroxide (150 mg) and ethanol (10 mL). The mixture was stirred under a hydrogen balloon for 48 h, filtered through Celite, and concentrated to yield the product (794 mg, 100%).
  • Examples 2-13 were prepared using methods analogous to those described for Example 1.
  • Examples 14-16 were prepared using methods analogous to those described for Example 1, substituting the reaction conditions of Cs 2 CO 3 in CH 3 CN at 75° C. for N,N-diisopropylethylamine in CH 3 CN at 50-70° C.
  • Examples 17-21 were prepared using methods analogous to those described for Example 1, substituting the reaction conditions of DMF at 50-80° C. for CH 3 CN at 70° C.
  • Examples 22-26 were prepared using methods analogous to those described for Example 1, substituting the reaction conditions of DMF at rt for CH 3 CN at 70° C.
  • Examples 27-42 were prepared using methods analogous to those described for Example 1, substituting DMF for CH 3 CN.
  • Examples 46-65 were prepared using methods analogous to those described for Example 45.
  • Examples 67-78 were prepared using methods analogous to those described for Example 45, substituting tert-amyl alcohol for CH 3 CN.
  • Examples 83-105 were prepared using methods analogous to those described for Example 82.
  • Examples 106-107 were prepared using methods analogous to those described for Example 82, substituting 50° C. for rt.
  • Examples 109-111 were prepared using methods analogous to those described for Example 82, substituting the following procedural differences: imine formation was conducted for 12 h at rt prior to the addition of sodium triacetoxyborohydride.
  • Examples 113-115 were prepared using methods analogous to those described for Example 112.
  • the title compound was synthesized as follows;
  • the salts were removed by filtration (at 70° C.) and the filter cake was washed with hot acetonitrile (5.0 kg). The filtrate was concentrated by distillation (at 50° C. in vacuo) of 8.1 kg solvent. The resulting suspension was cooled to 20° C. within 90 min, then to 0° C. in 2 h. After 2 h at 0° C., the product was isolated by centrifugation, washed with acetonitrile (5.66 kg) and dried in vacuo at 60° C. Yield: 1.696 kg dusky pink solid (74%).
  • succinate salts were prepared as follows. meso-N- ⁇ (3-endo)-8-[4-([1,3]Thiazolo[4,5-b]pyridin-2-yloxy)benzyl]-8-azabicyclo[3.2.1]oct-3-yl ⁇ acetamide (950 g, 2.08 mol) was dissolved in acetone (22.20 kg) at reflux. The red solution was filtered and stirred at 50-55° C. Succinic acid (261 g, 2.21 mol) was dissolved in acetone (3.905 kg) at 50° C.
  • salts of the above titled compound were prepared by adding malonic acid (1 equiv.) to the compound in methanol/methyl ethyl ketone; adding benzoic acid (1 equiv.) to the compound in methanol/acetonitrile; or adding succinic acid (1 equiv.) to the compound in acetone to achieve a malonate, benzoate, or succinate salt respectively.
  • malonic acid (1 equiv.)
  • benzoic acid (1 equiv.)
  • succinic acid (1 equiv.)
  • acetone to achieve a malonate, benzoate, or succinate salt respectively.
  • Some embodiments of compounds of this invention were prepared in the form of hydrochlorides, phosphates and sulfates, which were identified as solvated salts. Further embodiments were prepared in the form of fumarates,
  • Examples 119-128 were prepared using methods analogous to those described for Example 118.
  • the title compound was synthesized using the following procedure;
  • Step B 1- ⁇ (1S,4S)-5-[4-([1,3]Thiazolo[4,5-b]pyridin-2-yloxy)benzyl]-2,5-diazabicyclo[2.2.1]hept-2-yl ⁇ ethanone
  • Step C 1- ⁇ (1S,4S)-5-[4-([1,3]Thiazolo[4,5-b]pyridin-2-yloxy)benzyl]-2,5-diazabicyclo[2.2.1]hept-2-yl ⁇ ethanone Hydrochloride
  • the title compound was prepared in salt form, such as hydrochlorides, including hydrated hydrochlorides, such as the monohydrate.
  • Examples 129-130 were prepared using methods analogous to those described for Example 118, substituting Cs 2 CO 3 and the appropriate phenol for the sodium phenolate starting material.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Pulmonology (AREA)
  • Diabetes (AREA)
  • Dermatology (AREA)
  • Endocrinology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Rheumatology (AREA)
  • Neurology (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Hematology (AREA)
  • Biomedical Technology (AREA)
  • Urology & Nephrology (AREA)
  • Ophthalmology & Optometry (AREA)
  • Communicable Diseases (AREA)
  • Obesity (AREA)
  • Neurosurgery (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Pain & Pain Management (AREA)
  • Vascular Medicine (AREA)
  • Oncology (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Low-Molecular Organic Synthesis Reactions Using Catalysts (AREA)
US12/421,406 2008-04-11 2009-04-09 Thiazolopyridin-2-yloxy-phenyl and thiazolopyrazin-2-yloxy-phenyl amines as modulators of leukotriene A4 hydrolase Active US7939527B2 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US12/421,406 US7939527B2 (en) 2008-04-11 2009-04-09 Thiazolopyridin-2-yloxy-phenyl and thiazolopyrazin-2-yloxy-phenyl amines as modulators of leukotriene A4 hydrolase
US13/039,017 US8357684B2 (en) 2008-04-11 2011-03-02 Thyazolopyridin-2-yloxy-phenyl and thiazolopyrazin-2-yloxy-phenyl amines as modulators of leukotriene A4 hydrolase
US13/039,105 US20110190503A1 (en) 2008-04-11 2011-03-02 Thiazolopyridin-2-yloxy-phenyl and thiazolopyrazin-2-yloxy-phenyl amines as modulators of leukotriene A4 hydrolase

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US4434908P 2008-04-11 2008-04-11
US14912909P 2009-02-02 2009-02-02
US12/421,406 US7939527B2 (en) 2008-04-11 2009-04-09 Thiazolopyridin-2-yloxy-phenyl and thiazolopyrazin-2-yloxy-phenyl amines as modulators of leukotriene A4 hydrolase

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US13/039,105 Division US20110190503A1 (en) 2008-04-11 2011-03-02 Thiazolopyridin-2-yloxy-phenyl and thiazolopyrazin-2-yloxy-phenyl amines as modulators of leukotriene A4 hydrolase
US13/039,017 Continuation US8357684B2 (en) 2008-04-11 2011-03-02 Thyazolopyridin-2-yloxy-phenyl and thiazolopyrazin-2-yloxy-phenyl amines as modulators of leukotriene A4 hydrolase

Publications (2)

Publication Number Publication Date
US20090258854A1 US20090258854A1 (en) 2009-10-15
US7939527B2 true US7939527B2 (en) 2011-05-10

Family

ID=41130551

Family Applications (3)

Application Number Title Priority Date Filing Date
US12/421,406 Active US7939527B2 (en) 2008-04-11 2009-04-09 Thiazolopyridin-2-yloxy-phenyl and thiazolopyrazin-2-yloxy-phenyl amines as modulators of leukotriene A4 hydrolase
US13/039,105 Abandoned US20110190503A1 (en) 2008-04-11 2011-03-02 Thiazolopyridin-2-yloxy-phenyl and thiazolopyrazin-2-yloxy-phenyl amines as modulators of leukotriene A4 hydrolase
US13/039,017 Active US8357684B2 (en) 2008-04-11 2011-03-02 Thyazolopyridin-2-yloxy-phenyl and thiazolopyrazin-2-yloxy-phenyl amines as modulators of leukotriene A4 hydrolase

Family Applications After (2)

Application Number Title Priority Date Filing Date
US13/039,105 Abandoned US20110190503A1 (en) 2008-04-11 2011-03-02 Thiazolopyridin-2-yloxy-phenyl and thiazolopyrazin-2-yloxy-phenyl amines as modulators of leukotriene A4 hydrolase
US13/039,017 Active US8357684B2 (en) 2008-04-11 2011-03-02 Thyazolopyridin-2-yloxy-phenyl and thiazolopyrazin-2-yloxy-phenyl amines as modulators of leukotriene A4 hydrolase

Country Status (36)

Country Link
US (3) US7939527B2 (es)
EP (2) EP2336125B1 (es)
JP (2) JP5530423B2 (es)
KR (1) KR101228378B1 (es)
CN (1) CN102056933B (es)
AR (1) AR072353A1 (es)
AU (1) AU2009234140B2 (es)
BR (1) BRPI0911336A2 (es)
CA (1) CA2721099C (es)
CL (1) CL2009000877A1 (es)
CO (1) CO6260069A2 (es)
CY (1) CY1114628T1 (es)
DK (2) DK2268646T3 (es)
EA (1) EA018802B1 (es)
EC (1) ECSP10010558A (es)
ES (2) ES2440746T3 (es)
HK (2) HK1152041A1 (es)
HN (1) HN2010002074A (es)
HR (2) HRP20130278T1 (es)
IL (2) IL208480A (es)
JO (1) JO2911B1 (es)
MX (1) MX2010011154A (es)
MY (1) MY157597A (es)
NI (1) NI201000168A (es)
NZ (2) NZ588341A (es)
PE (1) PE20091779A1 (es)
PH (1) PH12013500094A1 (es)
PL (2) PL2336125T3 (es)
PT (2) PT2268646E (es)
RS (2) RS52712B (es)
SI (2) SI2336125T1 (es)
SV (1) SV2010003694A (es)
TW (2) TWI461196B (es)
UY (1) UY31760A (es)
WO (1) WO2009126806A2 (es)
ZA (1) ZA201008054B (es)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110159563A1 (en) * 2008-04-11 2011-06-30 Janssen Pharmaceutical Nv Thyazolopyridin-2-yloxy-phenyl and thiazolopyrazin-2-yloxy-phenyl amines as modulators of leukotriene A4 hydrolase

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2822942B1 (en) * 2012-03-06 2017-05-10 Boehringer Ingelheim International GmbH Benzodioxanes for inhibiting leukotriene production
EA201400976A1 (ru) 2012-03-06 2015-02-27 Бёрингер Ингельхайм Интернациональ Гмбх Бензодиоксаны в комбинации с другими активными средствами для ингибирования продуцирования лейкотриена
US9403830B2 (en) 2012-07-17 2016-08-02 Boehringer Ingelheim International Gmbh Inhibitors of leukotriene production
EP2746260A1 (en) 2012-12-21 2014-06-25 Basf Se Substituted [1,2,4]triazole and imidazole compounds
EP2746259A1 (en) 2012-12-21 2014-06-25 Basf Se Substituted [1,2,4]triazole and imidazole compounds
RU2686101C2 (ru) 2013-03-12 2019-04-24 Селтакссис, Инк. Способы ингибирования лейкотриен- а4-гидролазы
CA2906086A1 (en) * 2013-03-14 2014-09-25 Celtaxsys, Inc. Inhibitors of leukotriene a4 hydrolase
BR112015022226A2 (pt) 2013-03-14 2017-07-18 Celtaxsys Inc inibidores de leucotrieno a4 hidrolase
US10898484B2 (en) 2018-05-31 2021-01-26 Celltaxis, Llc Method of reducing pulmonary exacerbations in respiratory disease patients
BR112021013637A2 (pt) * 2019-01-11 2021-09-14 Naegis Pharmaceuticals Inc. Inibidores da síntese de leucotrieno

Citations (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2446823A1 (fr) 1979-01-16 1980-08-14 Delalande Sa Nouveaux derives du nor-tropane, leur procede de preparation et leur application en therapeutique
US4273778A (en) 1978-12-30 1981-06-16 Beecham Group, Limited Pharmaceutically active aza-bicyclo-benzamide derivatives
US4321378A (en) 1979-01-16 1982-03-23 Delalande S.A. 8-(5-Pyrimidinecarboxamide)nor-tropane derivatives
US4329466A (en) 1979-01-16 1982-05-11 Delalande S.A. Certain nor-tropan-8-amine-3-aryl or heteroaryl derivatives
US4352802A (en) 1980-06-10 1982-10-05 Beecham Group Limited Bicyclo[3.3.1]nonyl-benzamide
US4410535A (en) 1980-12-12 1983-10-18 Beecham Group Limited Aza-bicyclo-benzamide derivatives and pharmaceutical compositions and use thereof
US4424358A (en) 1979-01-16 1984-01-03 Delalande S.A. β-3-Amino nor-tropane compounds
US4432983A (en) 1981-04-30 1984-02-21 Research Corporation Conformationally restricted histamine H2 -receptor antagonists containing a tropane ring
US4471120A (en) 1979-01-16 1984-09-11 Delalande S.A. Nor-tropane derivatives
EP0266576A2 (en) 1986-10-08 1988-05-11 Bristol-Myers Squibb Company 1-Tert-alkyl-substituted naphthyridine and quinoline carboxylic acids as antibacterial agents
JPS63170356A (ja) 1986-12-30 1988-07-14 Yamanouchi Pharmaceut Co Ltd アニリン誘導体及びその製造法
EP0416521A1 (en) 1989-09-05 1991-03-13 G.D. Searle & Co. N-Benzyltropaneamides
EP0623621A1 (en) 1993-04-30 1994-11-09 Nisshin Flour Milling Co., Ltd. Oxazabicyclo derivatives and their use as 5-HT4 receptor agonists
WO1996011192A1 (en) 1994-10-11 1996-04-18 G.D. Searle & Co. Lta4 hydrolase inhibitors
WO1996041625A1 (en) 1995-06-12 1996-12-27 G.D. Searle & Co. Compositions comprising a cyclooxygenase-2 inhibitor and a leukotriene a4 hydrolase inhibitor
WO1997029774A1 (en) 1996-02-13 1997-08-21 G.D. Searle & Co. Combinations, having immunosuppressive effects, containing a cyclooxygenase-2 inhibitor and a leukotriene a4 hydrolase inhibitor
US5723492A (en) 1994-10-11 1998-03-03 G.D. Searle & Co. LTA4 hydrolase inhibitor pharmaceutical compositions and methods of use
US6110944A (en) 1997-03-12 2000-08-29 G. D. Searle & Co. LTA4, hydrolase inhibitors
WO2001081347A2 (en) 2000-04-27 2001-11-01 Abbott Laboratories Diazabicyclic central nervous system active agents
US6316490B1 (en) 1995-11-17 2001-11-13 Merck & Co., Inc. Substituted aryl compounds useful as modulators of acetylcholine receptors
US6432976B1 (en) 1999-10-29 2002-08-13 Merck & Co., Inc. 8-aza-bicyclo[3.2.1]octane NMDA/NR2B antagonists
US6559140B2 (en) 2000-03-09 2003-05-06 Abbott Laboratories Cyclic and bicyclic diamino histamine-3 receptor antagonists
WO2003037904A1 (fr) 2001-11-02 2003-05-08 Pierre Fabre Medicament NOUVEAUX DERIVES AMIDES HETEROAROMATIQUES DE 3β-AMINO AZABICYCLOOCTANE, LEUR PROCEDE DE PREPARATION ET LEURS APPLICATIONS EN THERAPEUTIQUE
US6632823B1 (en) 1997-12-22 2003-10-14 Merck & Co., Inc. Substituted pyridine compounds useful as modulators of acetylcholine receptors
WO2004103959A2 (en) 2003-05-16 2004-12-02 Ambit Biosciences Corporation Heterocyclic compounds and uses thereof
US20050043378A1 (en) 2003-07-28 2005-02-24 Axe Frank U. LTA4H modulators
WO2006002133A1 (en) 2004-06-22 2006-01-05 Schering Corporation Cannabinoid receptor ligands
WO2006004475A1 (en) 2004-06-30 2006-01-12 Autoliv Development Ab Arrangement for triggering a vehicle safety device
US20060074121A1 (en) 2004-03-03 2006-04-06 Chemocentryx, Inc. Bicyclic and bridged nitrogen heterocycles
US20060223792A1 (en) 2005-03-31 2006-10-05 Butler Christopher R Phenyl and pyridyl LTA4H modulators
WO2006133802A1 (en) 2005-06-14 2006-12-21 Laboratorios Almirall, S.A. N-amide derivatives of 8-azabicyclo[3.2.1]oct-3-yl as ccr1 antagonists
WO2007007069A1 (en) 2005-07-07 2007-01-18 Vernalis (R & D) Limited Azacyclic compounds as inhibitors of sensory neurone specific sodium channels
US20070079078A1 (en) 2002-05-29 2007-04-05 Takahiro Fujita Centralized storage management method
US20070155726A1 (en) 2005-12-29 2007-07-05 Schering Aktiengesellschaft Diamine derivatives as inhibitors of leukotriene A4 hydrolase
US20070167425A1 (en) 2003-08-29 2007-07-19 Shinji Nakade Compound capable of binding s1p receptor and pharmaceutical use thereof
WO2008016811A2 (en) 2006-07-31 2008-02-07 Neurogen Corporation Aminopiperidines and realted compounds
US20080057074A1 (en) 2004-09-13 2008-03-06 Yoshikazu Takaoka Nitrogenous Heterocylic Derivative and Medicine Containing the Same as an Active Ingredient
US20080194630A1 (en) 2007-02-14 2008-08-14 Barchuk William T LTA4H modulators and uses thereof
US20090111794A1 (en) * 2007-10-31 2009-04-30 Bacani Genesis M Aryl-substituted bridged or fused diamines as modulators of leukotriene A4 hydrolase

Family Cites Families (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5008A (en) * 1847-03-13 Machinery for cleaning
DE3137996A1 (de) * 1981-09-24 1983-04-28 Hoechst Ag, 6230 Frankfurt Heterocyclische phenylaether, verfahren zu deren herstellung und diese enthaltende herbizide mittel
US4571396A (en) * 1984-04-16 1986-02-18 Warner-Lambert Company Antibacterial agents
US4873346A (en) * 1985-09-20 1989-10-10 The Upjohn Company Substituted benzothiazoles, benzimidazoles, and benzoxazoles
DK171349B1 (da) * 1986-11-14 1996-09-16 Hoffmann La Roche Tetrahydronaphthalenderivater, fremgangsmåde til fremstilling deraf, lægemidler indeholdende forbindelserne samt anvendelse af forbindelserne til fremstilling af lægemidler
FI91859C (fi) * 1987-06-17 1994-08-25 Eisai Co Ltd Analogiamenetelmä antiallergisena aineena aktiivisen bentsotiatsolijohdannaisen valmistamiseksi
US5220063A (en) * 1991-05-10 1993-06-15 Hoechst Celanese Corporation Method for the preparation of arylalkanolacylamides
US5296486A (en) * 1991-09-24 1994-03-22 Boehringer Ingelheim Pharmaceuticals, Inc. Leukotriene biosynthesis inhibitors
US5472964A (en) * 1992-12-22 1995-12-05 Merck Frosst Canada, Inc. Diaryl 5,6-fused heterocyclic acids as leukotriene antagonists
US6632976B1 (en) * 1995-08-29 2003-10-14 Kirin Beer Kabushiki Kaisha Chimeric mice that are produced by microcell mediated chromosome transfer and that retain a human antibody gene
US6110994A (en) * 1996-06-14 2000-08-29 Cabot Corporation Polymeric products containing modified carbon products and methods of making and using the same
US7329682B2 (en) * 2003-04-03 2008-02-12 Ewha University-Industry Collaboration Foundation Method for inhibiting 5-lipoxygenase using a benzoxazole derivative
SE0301373D0 (sv) * 2003-05-09 2003-05-09 Astrazeneca Ab Novel compounds
TW200610761A (en) 2004-04-23 2006-04-01 Astrazeneca Ab Chemical compounds
US20090156465A1 (en) 2005-12-30 2009-06-18 Sattigeri Jitendra A Derivatives of beta-amino acid as dipeptidyl peptidase-iv inhibitors
EP2044038B1 (en) 2006-06-06 2014-07-02 Cornerstone Therapeutics Inc. Novel piperazines, pharmaceutical compositions and methods of use thereof
WO2008077103A1 (en) * 2006-12-19 2008-06-26 Auspex Pharmaceuticals, Inc. Preperation and utility of ccr5 inhibitors
ES2440746T3 (es) * 2008-04-11 2014-01-30 Janssen Pharmaceutica, N.V. Triazolopiridina-2-iloxi-fenil y triazolopiridina-iloxi-fenil amino como moduladores del leucotrieno A4 hidrolasa

Patent Citations (54)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4544660A (en) 1978-12-30 1985-10-01 Beecham Group P.L.C. Pharmaceutically active tropanes
US4336259A (en) 1978-12-30 1982-06-22 Beecham Group Limited Certain aza-bicyclo-benzamide derivatives and pharmaceutical compositions containing same
US4705858A (en) 1978-12-30 1987-11-10 Beecham Group P.L.C. Azabicyclo[3.3.1]nonanes
US4273778A (en) 1978-12-30 1981-06-16 Beecham Group, Limited Pharmaceutically active aza-bicyclo-benzamide derivatives
US4599420A (en) 1978-12-30 1986-07-08 Beecham Group Ltd. Azabicyclic intermediates for pharmaceutical compounds
US4321378A (en) 1979-01-16 1982-03-23 Delalande S.A. 8-(5-Pyrimidinecarboxamide)nor-tropane derivatives
US4471120A (en) 1979-01-16 1984-09-11 Delalande S.A. Nor-tropane derivatives
US4536580A (en) 1979-01-16 1985-08-20 Delalande S.A. 4-Amino-5-bromo-2-methoxy-N-[8-(benzyl)]-8-azabicyclo[3.2.1]-3-YL-benzamide or a pharmacologically acceptable acid addition salt thereof
US4424358A (en) 1979-01-16 1984-01-03 Delalande S.A. β-3-Amino nor-tropane compounds
US4329466A (en) 1979-01-16 1982-05-11 Delalande S.A. Certain nor-tropan-8-amine-3-aryl or heteroaryl derivatives
FR2446823A1 (fr) 1979-01-16 1980-08-14 Delalande Sa Nouveaux derives du nor-tropane, leur procede de preparation et leur application en therapeutique
US4352802A (en) 1980-06-10 1982-10-05 Beecham Group Limited Bicyclo[3.3.1]nonyl-benzamide
US4410535A (en) 1980-12-12 1983-10-18 Beecham Group Limited Aza-bicyclo-benzamide derivatives and pharmaceutical compositions and use thereof
US4432983A (en) 1981-04-30 1984-02-21 Research Corporation Conformationally restricted histamine H2 -receptor antagonists containing a tropane ring
EP0266576A2 (en) 1986-10-08 1988-05-11 Bristol-Myers Squibb Company 1-Tert-alkyl-substituted naphthyridine and quinoline carboxylic acids as antibacterial agents
JPS63170356A (ja) 1986-12-30 1988-07-14 Yamanouchi Pharmaceut Co Ltd アニリン誘導体及びその製造法
EP0416521A1 (en) 1989-09-05 1991-03-13 G.D. Searle & Co. N-Benzyltropaneamides
EP0623621A1 (en) 1993-04-30 1994-11-09 Nisshin Flour Milling Co., Ltd. Oxazabicyclo derivatives and their use as 5-HT4 receptor agonists
US5719306A (en) 1994-10-11 1998-02-17 G.D. Searle & Co. LTA4 hydrolase inhibitors
US5585492A (en) 1994-10-11 1996-12-17 G. D. Searle & Co. LTA4 Hydrolase inhibitors
WO1996011192A1 (en) 1994-10-11 1996-04-18 G.D. Searle & Co. Lta4 hydrolase inhibitors
US5723492A (en) 1994-10-11 1998-03-03 G.D. Searle & Co. LTA4 hydrolase inhibitor pharmaceutical compositions and methods of use
US6506876B1 (en) 1994-10-11 2003-01-14 G.D. Searle & Co. LTA4 hydrolase inhibitor pharmaceutical compositions and methods of use
WO1996041625A1 (en) 1995-06-12 1996-12-27 G.D. Searle & Co. Compositions comprising a cyclooxygenase-2 inhibitor and a leukotriene a4 hydrolase inhibitor
US5700816A (en) 1995-06-12 1997-12-23 Isakson; Peter C. Treatment of inflammation and inflammation-related disorders with a combination of a cyclooxygenase-2 inhibitor and a leukotriene A4 hydrolase inhibitor
US5990148A (en) 1995-06-12 1999-11-23 G.D. Searle & Co. Treatment of inflammation and inflammation-related disorders with a combination of a cyclooxygenase-2 inhibitor and a leukotriene A4 hydrolase inhibitor
US6316490B1 (en) 1995-11-17 2001-11-13 Merck & Co., Inc. Substituted aryl compounds useful as modulators of acetylcholine receptors
WO1997029774A1 (en) 1996-02-13 1997-08-21 G.D. Searle & Co. Combinations, having immunosuppressive effects, containing a cyclooxygenase-2 inhibitor and a leukotriene a4 hydrolase inhibitor
US20030004191A1 (en) 1996-02-13 2003-01-02 Pharmacia Corporation Immunosuppressive effects of administration of a cyclooxygenase-2 inhibitor and a leukotriene A4 hydrolase inhibitor
US20050043355A1 (en) 1996-02-13 2005-02-24 Pharmacia Corporation Immunosuppressive effects of administration of a cyclooxygenase-2 inhibitor and a leukotriene A4 hydrolase inhibitor
US6407140B1 (en) 1996-02-13 2002-06-18 G.D. Searle & Co. Immunosuppressive effects of administration of a cyclooxygenase-2 inhibitor and a leukotriene A4 hydrolase inhibitor
US6110944A (en) 1997-03-12 2000-08-29 G. D. Searle & Co. LTA4, hydrolase inhibitors
US6632823B1 (en) 1997-12-22 2003-10-14 Merck & Co., Inc. Substituted pyridine compounds useful as modulators of acetylcholine receptors
US6432976B1 (en) 1999-10-29 2002-08-13 Merck & Co., Inc. 8-aza-bicyclo[3.2.1]octane NMDA/NR2B antagonists
US6559140B2 (en) 2000-03-09 2003-05-06 Abbott Laboratories Cyclic and bicyclic diamino histamine-3 receptor antagonists
WO2001081347A2 (en) 2000-04-27 2001-11-01 Abbott Laboratories Diazabicyclic central nervous system active agents
WO2003037904A1 (fr) 2001-11-02 2003-05-08 Pierre Fabre Medicament NOUVEAUX DERIVES AMIDES HETEROAROMATIQUES DE 3β-AMINO AZABICYCLOOCTANE, LEUR PROCEDE DE PREPARATION ET LEURS APPLICATIONS EN THERAPEUTIQUE
US20070079078A1 (en) 2002-05-29 2007-04-05 Takahiro Fujita Centralized storage management method
WO2004103959A2 (en) 2003-05-16 2004-12-02 Ambit Biosciences Corporation Heterocyclic compounds and uses thereof
US20050043378A1 (en) 2003-07-28 2005-02-24 Axe Frank U. LTA4H modulators
US20050043379A1 (en) 2003-07-28 2005-02-24 Axe Frank U. LTA4H Modulators
US20070167425A1 (en) 2003-08-29 2007-07-19 Shinji Nakade Compound capable of binding s1p receptor and pharmaceutical use thereof
US20060074121A1 (en) 2004-03-03 2006-04-06 Chemocentryx, Inc. Bicyclic and bridged nitrogen heterocycles
WO2006002133A1 (en) 2004-06-22 2006-01-05 Schering Corporation Cannabinoid receptor ligands
WO2006004475A1 (en) 2004-06-30 2006-01-12 Autoliv Development Ab Arrangement for triggering a vehicle safety device
US20080057074A1 (en) 2004-09-13 2008-03-06 Yoshikazu Takaoka Nitrogenous Heterocylic Derivative and Medicine Containing the Same as an Active Ingredient
US20060223792A1 (en) 2005-03-31 2006-10-05 Butler Christopher R Phenyl and pyridyl LTA4H modulators
WO2006133802A1 (en) 2005-06-14 2006-12-21 Laboratorios Almirall, S.A. N-amide derivatives of 8-azabicyclo[3.2.1]oct-3-yl as ccr1 antagonists
WO2007007069A1 (en) 2005-07-07 2007-01-18 Vernalis (R & D) Limited Azacyclic compounds as inhibitors of sensory neurone specific sodium channels
US20070155726A1 (en) 2005-12-29 2007-07-05 Schering Aktiengesellschaft Diamine derivatives as inhibitors of leukotriene A4 hydrolase
WO2007079078A1 (en) 2005-12-29 2007-07-12 Bayer Schering Pharma Aktiengesellschaft Diamine derivatives as inhibitors of leukotriene a4 hydrolase
WO2008016811A2 (en) 2006-07-31 2008-02-07 Neurogen Corporation Aminopiperidines and realted compounds
US20080194630A1 (en) 2007-02-14 2008-08-14 Barchuk William T LTA4H modulators and uses thereof
US20090111794A1 (en) * 2007-10-31 2009-04-30 Bacani Genesis M Aryl-substituted bridged or fused diamines as modulators of leukotriene A4 hydrolase

Non-Patent Citations (123)

* Cited by examiner, † Cited by third party
Title
Agner, T. "Compliance Among Patients with Atopic Eczema", Acta Derm. Venereol. 2005; Suppl. 215; 33-35.
Ahluwalia et al., Inhibited Aortic Aneurysm Formation in BLT1-Deficient Mice, 2007, Journal of Immunology vol. 179:691-697.
Ahmadzadeh et al., "Relationship Betweetn Leukotriene B4 and Immunological Parameters in Rhematoid Synovial Fluids", 1991, Inflammation, vol. 15:497-503.
Alestas, T. et al. Enzymes involved in the biosynthesis of leukotriene B4 and prostaglandin E2 are active in sebaceous glands. J. Mol. Med. 2006, 84(1), 75-87.
Allen et al., "Inhaled Corticosteroids: Past Lessons and Future Issues" 2003 Allergy Clin. Immunol. vol. 112,3:S1-S40.
Andoh, T. et al. Intradermal leukotriene B4, but not prostaglandin E2, induces itch-associated responses in mice. Eur. J. Pharmacol. 1998, 353(1), 93-96.
Andoh, T. et al. Intradermal nociceptin elicits itch-associated responses through leukotriene B4 in mice. J. Investigativ. Dermatol. 2004, 123(1), 196-201.
Andoh, T. et al. Involvement of blockade of leukotriene B4 action in anti-pruritic effects of emedastine in mice. Eur. J. Pharmacol. 2000, 406(1), 149-152.
Andoh, T. et al. Involvement of leukotriene B4 in substance P-induced itch-associated response in mice. J. Investigativ. Dermatol. 2001, 117(6), 1621-1626.
Andoh, T. et al. Suppression by bepotastine besilate of substance P-induced itch-associated responses through the inhibition of the leukotriene B4 action in mice. Eur. J. Pharmacol. 2006, 547(1-3), 59-64.
Bagshawe et al. Antibody-Directed Enzyme Prodrug Therapy: A Review Drug Dev. Res. 1995, 34, 220-230.
Barnes, P.J. Future Advances in COPD Therapy. Respiration 2001, 68(5), 441-448.
Barone, F.C. et al. Time-related changes in myeloperoxidase activity and leukotriene B4 receptor binding reflect leukocyte influx in cerebral focal stroke. Mol. Chem. Neuropathol. 1995, 24(1), 13-30.
Bellamy et al., "Poor Perceptions and Expectations of Asthma Control: Results of the International Control of Asthma Symptoms (ICAS) Survey of Patients and General Practitioners" 2005, Primary Care Respiratory Journal, vol. 14:252-258.
Benoist, C. and D. Mathis. Mast Cells in Autoimmune Disease. Nature 2002, 420(6917), 875-878.
Berge et al. Pharmaceutical Salts Journal of Pharmaceutical Sciences 1977, 66(1), 1-19.
Bertolini et al. A New Rational Hypothesis for the Pharmacophore of the Active Metabolite of Leflunomide, A Potent Immunosuppressive Drug. J Med Chem 1997, 40, 2011-2016.
Biernacki, et al., "Increased Leukotriene B4 and 8-Isoprostane in Exhaled Breath Condensate of Patients with Exacerbations of COPD" 2003, Thorax.
Bodor et al. Novel Approaches to the Design of Safer Drugs: Soft Drugs and Site-Specific Chemical Delivery Systems Advances in Drug Research 1984, 13, 224-331.
Busse et al., "Advances in Immunology" 2001, New England Journal of Medicine, vol. 344:350-362.
Byrum, R.S. et al. Determination of the Contribution of Cysteinyl Leukotrienes and Leukotriene B4 in Acute Inflammatory Responses Using 5-Lipoxygenase- and Leukotriene A4 Hydrolase-Deficient Mice. J. Immunol. 1999, 163(12), 6810-6819.
Camp, R. et al. Production of Intraepidermal Microabscesses by Topical Application of Leukotriene B4. J. Invest. Dermatol. 1984, 82(2), 202-204.
Camp, R.D.R. et al. Responses of Human Skin to Intradermal Injection of Leukotrienes C4, D4and B4. Br. J. Pharmacol. 1983, 80(3), 497-502.
Carpagnano, G.E. et al. Increased leukotrien B4 and interleukin-6 in exhaled breath condensate in cystic fibrosis. Am. J. Respir. Crit. Care Med. 2003, 167(8), 1109-1112.
Chen, X. et al. Leukotriene A4 hydrolase as a target for cancer prevention and therapy. Curr. Cancer Drug Targets 2004, 4(3), 267-283.
Chen, X. et al. Leukotriene A4 hydrolase in rat and human esophageal adenocarcinomas and inhibitory effects of bestatin. J. Natl. Cancer Inst. 2003, 95(14), 1053-1061.
Cohen, J. The Immunopathogenesis of Sepsis. Nature (London) 2002, 420(6917), 885-891.
Collin, "Structural Requirements of D-2 Antidopaminergic Genzamides. Comparison with 5HT-3 Antiserotoninergic Orthopramides", 1991, Pharmacie de Belgique, vol. 46 Issue 1:55-66.
Coussens, L.M. et al. Inflammation and Cancer. Nature (London) 2002, 420(6917), 860-867.
Crooks, S.W. and R.A. Stockley. Leukotriene B4. Int. J. Biochem. Cell Biol. 1998, 30(2), 173-178.
Cunha, J.M. et al. The critical role of leukotriene B4 in antigen-induced mechanical hyperalgesia in immunised rats. Br. J. Pharmacol. 2003, 139(6), 1135-1145.
Dahlen et al., "Treatment of Asthma with Antileukotrienes: First Line or Last Resort Therapy" 2006, European Journal of Pharmacology, 533:40-56.
Dostert, P. et al., Studies on the Neuropleptic Benzamides. III-Synthesis and Antidopaminergic Properties of New 3-nortropane Derivatives, Eur. J. Med. Chem. vol. 19, No. 2, 1984, 105-110.
Ellis, C.N. et al. Cost of atopic dermatitis and eczema in the United States. J. Am. Acad. Dermatol. 2002, 46, 361-370.
Emingil, G. et al. Levels of leukotriene B4 in gingival crevicular fluid and gingival tissue in specific periodontal diseases. J. Periodontol. 2001, 72(8), 1025-1031.
Fitzgerald et al., "Asthma Control in Canada Remains Suboptimal: The Reality of Asthma Control (TRAC) Study" 2006, Can Resp. J. vol. 13, No. 5:253.
Fitzpatrick, F.A. et al. Effects of Leukotriene A4 on Neutrophil Activation. Ann. N. Y. Acad. Sci. 1994, 714, 64-74.
Fleisher, D. et al. Improved oral drug delivery: solubility limitations overcome by the use of prodrugs. Advanced Drug Delivery Reviews, 1996, 19, 115-130.
Ford-Hutchinson, A.W. et al. 5-Lipoxygenase. Ann. Rev. Biochem.1994, 63, 383-417.
Friedrich E.B. et al. Mechanisms of leukotriene B4-triggered monocyte adhesion. Arterioscler Thromb Vasc Biol 2003, 23, 1761-1767.
Frieri et al., "Allergen-Stimulated Leukotriene B4 and Interleukin-8 Levels in Patients with Asthma and Allergic Rhinitis-Modulation by a Lipid Pathway Inhibitor" 1998, Ann. Allergy Asthma Immunol. vol. 81:331.
Funk et al. Molecular Cloning and Amino Acid Sequence of Leukotriene A4 Hydrolase. PNAS 1987, 84, 6677-6681.
Gavezzotti, "Are Crystal Structures Predictable?"? Accounts of Chemical Research, 1994 vol. 27, 309-314.
Gelfand, E.W. et al. CD8+ T lymphocytes and leukotriene B4: novel interactions in the persistence and progression of asthma. J. Allergy Clin. Immunol. 2006, 117(3), 577-582.
Gierse et al. High level Experssion and Purification of Human Leukotriene A4 Hydrolase from Insect Cells Infected with a Baculovirus Vector. Protein Expression and Purification, 1993, 4, 358-366.
Gompertz, S. et al. Changes in bronchial inflammation during acute exacerbations of chronic bronchitis. Eur. Respir. J. 2001, 17(6), 1112-1119.
Goodarzi, K. et al. Leukotriene B4 and BLT1 Control Cytotoxic Effector T Cell Recruitment to Inflamed Tissues. Nat. Immunol. 2003) 4(10), 965-973.
Grice et al., Discovery of Potent and Selective Leukotriene A4 Hydrolase Inhibitors Abstracts of Papers, 234th ACS National Meeting, Boston, MA, US Aug. 19, 2007.
Griffiths, R.J. et al. Leukotriene B4 Plays a Critical Role in the Progression of Collagen-Induced Arthritis. PNAS 1995, 92(2), 517-521.
Hakonarson, H. et al. Effects of a 5-lipoxygenase-activating protein inhibitor on biomarkers associated with risk of myocardial infarction. A randomized trial. JAMA 2005, 293(18), 2245-2256.
Hanifin, J.M. et al. Guidelines of care for atopic dermatitis, developed in accordance with the American Academy of Dermatology (AAD)/American Academy of Dermatology Association Administrative Regulations for Evidence-Based Clinical Practice Guidelines. J. Am. Acad. Dermatol. 2004, 50, 391-404.
Helgadottir A. et al. A variant of the gene encoding leukotriene A4 hydrolase confers ethnicity-specific risk of myocardial infarction. Nat Genet. 2006, 38, 68-74.
Helgadottir, A. et al. The Gene Encoding 5-Lipoxygenase Activating Protein Confers Risk of Myocardial Infarction and Stroke. Nat. Genet. 2004, 36(3), 233-239.
Huang L, et al. Molecular and Biological Characterization of the Murine Leukotriene B4 Receptor Expressed on Eosinophils. J. Exp. Med. 1998, 188(6), 1063-1074.
Hwang, S.W. et al. Direct activation of capsaicin receptors by products of lipoxygenases: endogenous capsaicin-like substances. Proc. Natl. Acad. Sci. USA 2000, 97(11), 6155-6160.
Ikai, K. Psoriasis and the Arachidonic Acid Cascade, Jour. Of Derm. Sci., 1999, 21, 135-146.
International Search Report dated Jan. 22, 2010 for International Application No. PCT/US2009/040070.
Jala, V.R. et al. Leukotrienes and Atherosclerosis: New Role for Old Mediators. Trends Immunol. 2004, 25(6), 315-322.
Kachur, J.F. et al. Pharmacological Characterization of SC-57461A (3-[Methyl[3-[4-(phenylmethyl)phenoxy]propyl]amino]propanoic acid HCI), a Potent and Selective Inhibitor of Leukotriene A4 Hydrolase II: In Vivo Studies. J. Pharmacol. Exp. Ther. 2002, 300(2), 583-587.
Klein, A. et al. Stem Cell Factor Plays a Major Role in the Recruitment of Eosinophils in Allergic Pleurisy in Mice via the Production of Leukotriene B4. J. Immunol. 2000, 164(8), 4271-4276.
Koro et al., Chemical Mediators in Atopic Dermatitis: Involvement of Leukotriene B4 Released by a Type I Allergic Reaction in the Pathogenesis of topic Dermatitis, J. Allergy Clin. Immunol. (1999) vol. 103:663-670.
Laughter, D. et al. The prevalence of atopic dermatitis in Oregon schoolchildren. J. Am. Acad. Dermatol. 2000, 43, 649-655.
Liao, T. et al. Blockade of the interaction of leukotriene B4 with its receptor prevents development of autoimmune uveitis. Invest. Ophthalmol. Vis. Sci. 2006, 47(4), 1543-1549.
Libby, P. Inflammation in Atherosclerosis. Nature (London) 2002, 420(6917), 868-874.
Lundeen et al., "Leukotriene B4 Receptors BLT1 and BLT2: Expression and Function in Human and Murine Mast Cells" 2006, The Journal of Immunology, vol. 177:3439-3447.
Mathis et al., "Role of Leukotriene B4 Receptors iin Rheumatoid Arthritis" 2007 Autoimmunity Reviews, vol. 7:12-17.
Message et al., "The Immunology of Virus Infection in Asthma" 2001, European Respiratory Journal, vol. 18:1013-1025.
Milgrom et al., "Noncompliance and Treatment Failure in Children with Asthma" 1996, J. Allergy Clin. Immunol. vol. 98:1051-1057.
Miyahara N. et al. Role of the LTB4/BLT1 pathway in allergen-induced airway hyperresponsiveness and inflammation. Allergy Intl. 2006, 55(2), 91-97.
Moore et al., "Severe Asthma: An Overview" 2006 American Academy of Allergy, Asthma and Immunology, vol. 117:487-494.
Morisseau et al., "Potent Urea and Carbamate Inhibitors of Soluble Epoxide Hydrolases" Proceedings of the National Academy of Sciences 1999, 96, 8849-8854.
Munafo, D.A. et al. Leukotriene A4 Hydrolase in Human Bronchoalveolar Lavage Fluid. J. Clin. Invest. 1994, 93(3), 1042-1050.
Nakae, H. et al. Relationship between cytokines and leukotriene B4 in sepsis. Res. Commun. Chem. Pathol. Pharmacol. 1994, 83(2), 151-156.
Nathan, C. Points of Control in Inflammation. Nature (London) 2002, 420(6917), 846-852.
Neu et al., "Leukotrienes in Patients with Clinically Active Multiple Sclerosis", 2002 Acta Neurol. Scand, vol. 105:63-66.
Ott, V.L. et al. Cell-Dependent Migration of Effector CD8+ T Cells Through Production of Leukotriene B4. Nat. Immunol. 2003, 4(10), 974-981.
Patani et al.: "Biosoterism: A Rational Approach in Drug Design" 1996; Chem. Rev.; 96:3147-3176.
Penning et al., "Pyrrolidine and Piperidine Analogues of SC-57461A as Potent, orally Active Inhibitors of Leukotriene A4 Hydrolase" Bioorganic & Medicinal Chemistry Letters 2002, 12, 3383-3386.
Penning, T.D. Inhibitor of Leukotriene A4 (LTA4) Hydrolase as Potential Anti-Inflammatory Agents. Curr. Pharm. Des. 2001, 7(3), 163-179.
Rabier, et al., "Neuropeptides Modulate Leukotriene B4 Mitogenicity Toward Cultured Human Keratinocytes", 1993 J. Invest. Deermatol. vol. 110:132-136.
Rao et al., "Anti-Inflammatory Activity of A potent, Selective Leukotriene A4 Hydrolase Inhibitor in Comparison with the 5-Lipoxygenase Inhibitor Zileuton", 2007, vol. 321, No. 3:1154-1160.
Reid, G.K. et al. Correlation Between Expression of 5-Lipoxygenase-Activating Protein, 5-Lipoxygenase, and Cellular Leukotriene Synthesis. J. Biol. Chem. 1990, 265(32), 19818-19823.
Robinson, R. et al. Discovery of the Hemifumarate and (alpha-L-Alanyloxy) methyl Ether as Prodrugs of an Antirheumatic Oxindole: Prodrugs for the Enolic OH Group. J. Med Chem. 1996, 39, 10-18.
Robinson, R. et al. Discovery of the Hemifumarate and (α-L-Alanyloxy) methyl Ether as Prodrugs of an Antirheumatic Oxindole: Prodrugs for the Enolic OH Group. J. Med Chem. 1996, 39, 10-18.
Ruzicka et al., "Skin Levels of Arachidonic Acid-Derived Inflammatory Mediators and Histamine in Atopic Dermatitis and Psoriasis" 1986, J. Invest. Dermatol. vol. 86:105-108.
Samuelsson, B et al. Enzymes Involved in the Biosynthesis of Leukotriene B4. J. Biol. Chem. 1989, 264(33), 19469-19472.
Samuelsson, B. et al. Leukotrienes: Mediators of Immediate Hypersensitivity Reactions and Inflammation. Science (Washington, D.C.) 1983, 220 (4597), 568-575.
Schimmer et al., Adrenocorticotropic Hormone; Adrenocortical Steroids and Their Synthetic Analogs; Inhibitors of the Synthesis and Action of Adrenocortical Hormones 2001; in Hardman JG, Limbird LE; eds. Goodman and Gilman's The Pharmacological Basis of Therapeutics, 10th ed. New York: McGraw-Hill; 1666-1668.
Seggev et al., "Serum Leukotriene B4 Levels in Patients with Obstructive Pulmonary Disease" 1991, Chest. vol. 99-289-291.
Shan et al. Prodrug strategies based on intramolecular cyclization reactions. Journal of Pharmaceutical Sciences 1997, 86(7), 765-767.
Sharon, P. et al. Enhanced Synthesis of Leukotriene B4 by Colonic Mucosa in Inflammatory Bowel Disease. Gastroenterology 1984, 86(3), 453-460.
Sidbury, R. et al. Old, new, and emerging therapies for atopic dermatitis. Dermatol. Clin. 2000, 18(1), 1-11.
Steinberg, D. Atherogenesis in Perspective: Hypercholesterolemia and Inflammation as Partners in Crime. Nat. Med. 2002, 8(11), 1211-1217.
Su, J.C. et al. Atopic eczema: its impact on the family and financial cost. Arch. Dis. Child 1997, 76, 159-162.
Subbarao, K. et al. Role of Leukotriene B4 Receptors in the Development of Atherosclerosis: Potential Mechanisms. Arterioscler. Thromb. Vasc. Biol. 2004, 24, 369-375.
Suzuki et al., A Practical Procedure for Preparation of N-(endo-8-(3-Hydroxy)Propyl-8-Azabicyclo[3.2.1.]Oct-3-YL)-1-Isopropyl-2-Oxo-1,2-Dihydro-3-Quinoline-Carboxamide (TS-951) 2000, Heterocycles, vol. 53, No. 11 pp. 2471-2485.
Szefler et al., Am. J. Respir. Crit. Care Med. Feb. 1, 2003; 176(3):290-291.
Tager, A.M. et al. Leukotriene B4 Receptor BLT1 Mediates Early Effector T Cell Recruitment. Nat. Immunol. 2003, 4(10), 982-990.
Takakuwa, T. et al. Relationships between plasma levels of type-II phospholipase A2, PAF-acetylhydrolase, leukotriene B4, complements, endothelin-1 and thrombomodulin in patients with sepsis. Res. Commun. Chem. Pathol. Pharmacol. 1994, 84(3), 271-281.
Terawaki K. et al. Absence of leukotriene B4 receptor 1 confers resistance to airway hyperresponsiveness and Th2-type immune responses. J Immunol. 2005, 17(7), 4217-4225.
Tracey, K.J. The Inflammatory Reflex. Nature (London) 2002, 420(6917), 853-859.
Tsuji F. et al. Involvement of Leukotriene B4 in Arthritis Models. Life Sci. 1998, 64(3), L51-L56.
Turner et al., "In Vitro and In Vivo Effects of Leukotriene B4 Antagonism in a Primate Model of Asthma" 1996, J. Clin. Invest., vol. 97-381-387.
Vippagunta et al., "Crystalline Solids", Advanced Drug Delivery Reviews, 2001 vol. 48, 3-26.
Wang, S. et al. A Novel Hepatointestinal Leukotriene B4 Receptor. Cloning and Functional Characterization. J. Biol. Chem. 2000, 275(52), 40686-40694.
Wedi et al., Pathophysiological Role of Leukotrienes in Dermatological Diseases, 2001, BioDrugs vol. 15(11):729-743.
Weiner, H.L., et al. Inflammation and Therapeutic Vaccination in CNS Diseases. Nature (London) 2002, 420(6917), 879-884.
Wenzel et al., "Bronchoscopic Evaluation of Severe Asthma" 1997, American Journal of Respiratory and Critical Care Medicine, vol. 156:737-743.
West, Anthony R., Solid State Chemistry and its Applications, Wiley, New York, 1988, pp. 358 &35565.
Whatling et al., "The Potential Link Between Atherosclerosis and the 5-Lipoxygenase Pathway Investigational Agents with New Implications for the Cardiovascular Field" 2007, Informa Healthcare, vol. 16:1879-1893.
Whittle et al., "Attentuation of Inflammation and Cytokine Production in Rat Colitis by a Novel Selective Inhibitor of Leukotriene A4 Hydrolase" 2008, British Journal of Pharmacology, vol. 153:983-991.
Willemsen et al., "Deffective Metabolism of Leukotriene B4 in the Sjogren-Larsson Syndrome" 2001, Journal of the Neurological Sciences vol. 183:61-67.
Willemsen M.A. et al. Clinical and biochemical effects of zileuton in patients with the Sjogren-Larsson syndrome. Eur J Pediatr. 2001,160, 711-717.
Wolff, M.E. Burger's Medicinal Chemistry 4th Ed. Par I, Wiley: New York, 1979 336-337.
Woodmansee D.P. et al. Simon RA. A pilot study examining the role of zileuton in atopic dermatitis. Ann Allergy Asthma Immunol. 1999, 83, 548-552.
Yokomizo, T. et al. A Second Leukotriene B4 Receptor, BLT2: A New Therapeutic Target in Inflammation and Immunological Disorders. J. Exp. Med. 2000, 192(3), 421-431.
Yokomizo, T. et al. Co-expression of two LTB4 Receptors in Human Mononuclear Cells. Life Sci. 2001, 68(19-20), 2207-2212.
Yokomizo, T. et al. Leukotriene A4 Hydrolase and Leukotriene B4 Metabolism. J. Lipid Mediat. Cell Signal. 1995, 12(2-3), 321-332.
Yokomizo, T. et al. Leukotriene B4: Metabolism and Signal Transduction. Arch. Biochem. Biophys. 2001, 385(2), 231-241.
Zhu, L. et al. A convenient synthesis of 2-mercapto and 2-chiorobenzotniazoles. J. Heterocyclic Chem. 2005, 42, 727-730.
Zhu, Y.I. et al. Preview of Potential Therapeutic Applications of Leukotriene B4 Inhibitors in Dermatology. Skin Pharmacol. Appl. Skin Physiol. 2000, 13(5), 235-245.
Zouboulis, Ch.C. et al. A new concept for acne therapy: a pilot study with zileuton, an oral 5-lipoxygenase inhibitor. Arch. Dermatol. 2003, 139(5), 668-670.
Zouboulis, Ch.C. et al. Zileuton, an Oral 5-Lipoxygenase Inhibitor, Directly Reduces Sebum Production. Dermatology 2005, 210(1), 36-38.

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110159563A1 (en) * 2008-04-11 2011-06-30 Janssen Pharmaceutical Nv Thyazolopyridin-2-yloxy-phenyl and thiazolopyrazin-2-yloxy-phenyl amines as modulators of leukotriene A4 hydrolase
US8357684B2 (en) 2008-04-11 2013-01-22 Janssen Pharmaceutica Nv Thyazolopyridin-2-yloxy-phenyl and thiazolopyrazin-2-yloxy-phenyl amines as modulators of leukotriene A4 hydrolase

Also Published As

Publication number Publication date
WO2009126806A8 (en) 2010-11-18
TW201002320A (en) 2010-01-16
HRP20131192T1 (hr) 2014-01-17
SV2010003694A (es) 2011-03-17
DK2336125T3 (da) 2013-04-08
PL2336125T3 (pl) 2013-06-28
PT2268646E (pt) 2013-12-10
EP2268646B1 (en) 2013-10-02
TWI461196B (zh) 2014-11-21
JP5530423B2 (ja) 2014-06-25
IL226244A (en) 2014-12-31
HRP20130278T1 (en) 2013-06-30
IL226244A0 (en) 2013-06-27
JP2014144949A (ja) 2014-08-14
WO2009126806A3 (en) 2010-03-18
RS52712B (en) 2013-08-30
KR101228378B1 (ko) 2013-01-31
US20110190503A1 (en) 2011-08-04
UY31760A (es) 2009-11-10
RS53081B (en) 2014-06-30
JO2911B1 (en) 2015-09-15
JP5675943B2 (ja) 2015-02-25
ES2400875T3 (es) 2013-04-15
AU2009234140A1 (en) 2009-10-15
CA2721099C (en) 2017-04-25
ECSP10010558A (es) 2010-11-30
EP2268646B9 (en) 2014-11-19
JP2011516571A (ja) 2011-05-26
ZA201008054B (en) 2012-04-25
CA2721099A1 (en) 2009-10-15
DK2268646T3 (da) 2013-11-18
PE20091779A1 (es) 2009-11-14
NZ588341A (en) 2012-06-29
HK1152041A1 (en) 2012-02-17
CN102056933B (zh) 2014-09-17
EP2336125B1 (en) 2013-01-09
PH12013500094B1 (en) 2015-02-23
AU2009234140B2 (en) 2013-04-11
CO6260069A2 (es) 2011-03-22
CY1114628T1 (el) 2016-10-05
US8357684B2 (en) 2013-01-22
EP2268646A2 (en) 2011-01-05
PT2336125E (pt) 2013-03-18
HN2010002074A (es) 2012-12-03
EA201071186A1 (ru) 2011-06-30
EA018802B1 (ru) 2013-10-30
IL208480A0 (en) 2010-12-30
KR20110013386A (ko) 2011-02-09
WO2009126806A2 (en) 2009-10-15
ES2440746T3 (es) 2014-01-30
HK1158208A1 (en) 2012-07-13
EP2336125A1 (en) 2011-06-22
NI201000168A (es) 2015-05-27
NZ599853A (en) 2012-08-31
SI2336125T1 (sl) 2013-04-30
BRPI0911336A2 (pt) 2021-06-29
SI2268646T1 (sl) 2013-12-31
PL2268646T3 (pl) 2014-02-28
IL208480A (en) 2014-11-30
TW201420100A (zh) 2014-06-01
CN102056933A (zh) 2011-05-11
MX2010011154A (es) 2010-12-21
PH12013500094A1 (en) 2015-02-23
AR072353A1 (es) 2010-08-25
US20110159563A1 (en) 2011-06-30
CL2009000877A1 (es) 2009-11-27
MY157597A (en) 2016-06-30
US20090258854A1 (en) 2009-10-15

Similar Documents

Publication Publication Date Title
US7939527B2 (en) Thiazolopyridin-2-yloxy-phenyl and thiazolopyrazin-2-yloxy-phenyl amines as modulators of leukotriene A4 hydrolase
US7935725B2 (en) Aryl-substituted bridged or fused diamines as modulators of leukotriene A4 hydrolase
US8399465B2 (en) Compounds with two fused bicyclic heteroaryl moieties as modulators of leukotriene A4 hydrolase
AU2013205607B2 (en) Thiazolopyridin-2-yloxy-phenyl and thiazolopyrazin-2-yloxy-phenyl amines as modulators of leukotriene A4 hydrolase

Legal Events

Date Code Title Description
AS Assignment

Owner name: JANSSEN PHARMACEUTICA NV, BELGIUM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:FOURIE, ANNE M.;REEL/FRAME:022603/0799

Effective date: 20090406

Owner name: JANSSEN PHARMACEUTICA NV, BELGIUM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:WICKBOLDT, ALVAH T.;REEL/FRAME:022603/0960

Effective date: 20090402

Owner name: JANSSEN PHARMACEUTICA NV, BELGIUM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:GRICE, CHERYL A.;REEL/FRAME:022603/0964

Effective date: 20090327

Owner name: JANSSEN PHARMACEUTICA NV, BELGIUM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:CHEUNG, EUGENE Y.;REEL/FRAME:022606/0075

Effective date: 20090327

Owner name: JANSSEN PHARMACEUTICA NV, BELGIUM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BROGGINI, DIEGO;LOCHNER, SUSANNE;ZINSER, HARTMUT;REEL/FRAME:022606/0845

Effective date: 20090403

Owner name: JANSSEN PHARMACEUTICA NV, BELGIUM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BACANI, GENESIS M.;CHROVIAN, CHRISTA C.;DENG, XIAOHU;AND OTHERS;REEL/FRAME:022606/0850;SIGNING DATES FROM 20090330 TO 20090331

Owner name: JANSSEN PHARMACEUTICA NV, BELGIUM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BACANI, GENESIS M.;CHROVIAN, CHRISTA C.;DENG, XIAOHU;AND OTHERS;SIGNING DATES FROM 20090330 TO 20090331;REEL/FRAME:022606/0850

STCF Information on status: patent grant

Free format text: PATENTED CASE

CC Certificate of correction
CC Certificate of correction
FPAY Fee payment

Year of fee payment: 4

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 8TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1552); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 8

FEPP Fee payment procedure

Free format text: MAINTENANCE FEE REMINDER MAILED (ORIGINAL EVENT CODE: REM.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

LAPS Lapse for failure to pay maintenance fees

Free format text: PATENT EXPIRED FOR FAILURE TO PAY MAINTENANCE FEES (ORIGINAL EVENT CODE: EXP.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20230510